ArticlePDF AvailableLiterature Review

Small bowel transplant - novel indications and recent progress

Taylor & Francis
Expert Review of Gastroenterology & Hepatology
Authors:

Abstract

Introduction: Advances in the management of intestinal failure have led to a reduction in the number of intestinal transplants. The number of bowel transplants has been mainly stable even though a slight increase has been observed in the last five years. Areas covered: Standard indication includes patients with a reasonable life expectancy. Recent progress can be deduced by the increased number of intestine transplants in adults: this is due to the continuous improvement of 1- year graft survival worldwide (without differences in 3- and 5-year) associated with better abdominal wall closure techniques. This review aims to provide an update on new indications and changes in trends of pediatric and adult intestine transplantation. This analysis, which stretches through the past 5 years, is based on a collection of related manuscripts from PubMed. Expert commentary: Intestinal transplants should be solely intended for a group of individuals for whom indications for transplantation are clear and both medical and surgical rehabilitations have failed. Nevertheless, the many protocols developed over the years have not yet solved the key question represented by the over-immunosuppression. Novel indications and recent progress in the bowel transplant field, minimal yet consistent, represent a pathway to be followed.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ierh20
Expert Review of Gastroenterology & Hepatology
ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ierh20
Small bowel transplant – novel indications and
recent progress
Noemi Zorzetti, Ignazio Roberto Marino, Salvatore Sorrenti, Giuseppe
Giovanni Navarra, Vito D’Andrea & Augusto Lauro
To cite this article: Noemi Zorzetti, Ignazio Roberto Marino, Salvatore Sorrenti, Giuseppe
Giovanni Navarra, Vito D’Andrea & Augusto Lauro (2023) Small bowel transplant – novel
indications and recent progress, Expert Review of Gastroenterology & Hepatology, 17:7,
677-690, DOI: 10.1080/17474124.2023.2221433
To link to this article: https://doi.org/10.1080/17474124.2023.2221433
Published online: 05 Jun 2023.
Submit your article to this journal
Article views: 40
View related articles
View Crossmark data
REVIEW
Small bowel transplant novel indications and recent progress
Noemi Zorzetti
a,b
, Ignazio Roberto Marino
c
, Salvatore Sorrenti
b
, Giuseppe Giovanni Navarra
a
, Vito D’Andrea
b
and Augusto Lauro
b
a
General Surgery, Ospedale Civile “A. Costa”, Alto Reno Terme, Bologna, Italy;
b
Department of Surgical Sciences, Sapienza University of Rome,
Rome, Italy;
c
Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
ABSTRACT
Introduction: Advances in the management of intestinal failure have led to a reduction in the number
of intestinal transplants. The number of bowel transplants has been mainly stable even though a slight
increase has been observed in the last 5 years.
Areas covered: Standard indication includes patients with a reasonable life expectancy. Recent pro-
gress can be deduced by the increased number of intestine transplants in adults: this is due to the
continuous improvement of 1-year graft survival worldwide (without differences in 3- and 5-year)
associated with better abdominal wall closure techniques. This review aims to provide an update on
new indications and changes in trends of pediatric and adult intestine transplantation. This analysis,
which stretches through the past 5 years, is based on a collection of related manuscripts from PubMed.
Expert commentary: Intestinal transplants should be solely intended for a group of individuals for
whom indications for transplantation are clear and both medical and surgical rehabilitations have failed.
Nevertheless, many protocols developed over the years have not yet solved the key question repre-
sented by the over-immunosuppression. Novel indications and recent progress in the bowel transplant
field, minimal yet consistent, represent a pathway to be followed.
ARTICLE HISTORY
Received 12 December 2022
Accepted 24 May 2023
KEYWORDS
Intestinal transplantation;
intestinal failure; short
bowel syndrome; home
parenteral nutrition; enteral
autonomy; quality of life
1. Introduction: intestinal failure and short bowel
syndrome
Intestinal failure (IF) is defined as any cause of gastrointestinal
(GI) dysfunction that results in the inability to meet nutritional
demands, necessitating either temporary or indefinite depen-
dence on parenteral nutrition (PN) [1].
IF is mainly due to extensive surgical resection that leads to
short bowel syndrome (SBS) [2] which represents a severe and
rare clinical condition defined by malabsorption with conse-
quent malnutrition, anemia, and weight loss. Different etiolo-
gies, of new onset or chronic ones, are widely described in
literature among adults and the pediatric population [3,4]
Table 1.
Different phases of SBS have been recognized (acute phase,
adaptation phase, and chronic phase). Generally, the reversibil-
ity of this condition could easily appear when there is one of
the following situations:
more than 35 cm of small bowel along with a jejunoileal
anastomosis and an intact ileocecal valve and colon;
more than 60 cm of small bowel with a jejuno-colonic
anastomosis;
more than 115 cm of small bowel with an end
jejunostomy.
The final phase is the maintenance one, characterized by
special diets, supplementation of nutrients, and pharmacolo-
gical treatments [5].
Furthermore, SBS represents nowadays a new frontier in
biomarker research (especially growth factors and citrulline)
for diagnostic, prognostic, or predictive uses. The introduction
of biomarker research into a medical routine is expected to
represent helpful clinical support for patients, their families,
and healthcare systems [6]. However, these tests are still not
widely accepted and not available in all centers, therefore
limiting their clinical utility [7].
Intestinal adaptation (IA) is a response to the loss of the
small intestine and it is essential to restore enteral autonomy
after massive small bowel resection (MSBR): multidisciplinary
management of SBS could allow the patient to achieve nutri-
tional autonomy. In a well-selected population, specific surgical
lengthening procedures plus intestinal medical rehabilitation,
together with novel approaches (such as manipulating micro-
biota or tissue bioengineering), could also be attempted to
wean HPN off [8], while intestinal transplantation (ITx) is
reserved for highly selected patients.
A significant portion of patients are being referred for HPN
as a bridge to reconstructive surgery, and this requires the
close involvement of gastrointestinal surgeons in HPN teams
[9]. Furthermore, the increase in numbers, age, and turnover
of patients dependent on HPN requires more resources and
nutrition teams; the use of technology for remote consulta-
tions can improve the efficiency of clinics and avoid some
patients to travel [9].
If this goal is not achieved, chronic IF appears, with nutrient
deficiencies and other organ-specific complications [Table 2].
CONTACT Augusto Lauro augusto.lauro@uniroma1.it Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY
2023, VOL. 17, NO. 7, 677–690
https://doi.org/10.1080/17474124.2023.2221433
© 2023 Informa UK Limited, trading as Taylor & Francis Group
The first-line therapy for IF remains home PN (HPN) and
frequent monitoring. Although this therapy is reliable, it is not
without complications, such as venous thrombosis and IF-
associated liver disease (IFALD), or increased morbidity related
to catheter-associated infections [10–13]. Overall, in both the
adult and pediatric populations, HPN shows a 76% survival
rate at 5 years [14,15].
If we take into consideration the global increase of
patients that undergo HPN (both to support IA in SBS or to
treat IF), their heterogeneity and the relatively few locations
of the specialized centers, we can see how telemedicine
could today become a very helpful tool for these patients.
The possibility of a video call service, alongside with in-
person consultations and visits, could provide not only
a strategy to reduce patients’ need to travel, bettering their
quality of life (QOL) and reducing the economical and geo-
graphical barriers, but also allows equal access to high-
quality care [16,17].
Lastly, as the number of elderly patients that survive the
once fatal illnesses grows, the need for a new at-home or
residential care-supporting model service becomes more and
more crucial [18–20].
2. Timing and extension of small bowel resection:
considerations for general management
The functional consequences of SBS depend on multifactorial
issues such as age, timing, patient’s underlying diagnosis,
overall health and the extension of resection such as the
intestinal tract involved [21]. At birth, the small bowel length
(SBL) of full-term newborns is 250 cm, and it then lengthens
during the first year. One of the main predictive factors for
intestinal rehabilitation after MSBR in the pediatric population
is a residual SBL > 25% of the expected gestational age [22]
while, in the adult population, a residual SBL > 67% of the
normal length (that corresponds to approximately 200 cm) is
the minimum value according to international literature [23].
Furthermore, two other substantial issues have to be consid-
ered for enteral autonomy: the first is the importance of the
colon in continuity with the intestine after MSBR, with its
energy-absorptive function; in fact, half of the colon and 50
cm of small bowel can be considered equal in promoting the
digestive process [3]. The second one is the difference of roles
in digestion in jejunum and ileum: IA is a natural compensa-
tory process but ileum has a better adaptive potential than
jejunum and remnant ileum (above all if in continuity with the
colon) can guarantee a better absorption and a possible faster
weaning from HPN [8,24].
Thus, taking into account all these different aspects, recent
studies [25] have suggested that 75 cm could be the minimum
length of remnant small bowel to obtain nutritional autonomy
after medical rehabilitation in adult patients when the colon
or part of it is present.
Nevertheless, apart from the underlying cause of SBS, the
outcome of enteral autonomy depends on the type of remnant
bowel, the presence of a stoma or an anastomosis, full or partial
presence of the colon, the presence of ileocecal valve (ICV) and
the patient’s age plus his/her general conditions [26,27].
3. Medical approach to short bowel syndrome
At present, one of the main treatment options for SBS is
medical intestinal rehabilitation. In order to improve survival,
Article highlights
Intestinal transplantation should be strictly reserved to a group of
individuals for whom the indications to transplant are clear and
medical and surgical rehabilitations have failed.
Medical approaches (such as rehabilitation or in the near future
proteomic therapy) and non-transplant surgical techniques have
a central role in the management of patients with intestinal failure.
Novel criteria for intestinal transplant are recently emerged imple-
menting new indications to transplant apart from the standard ones.
Quality of life itself does not represent an option for transplantation
unless other indications are present.
The intestine is the most immunologically complex solid organ
allograft with the greatest risk of both rejection and graft-versus-
host disease: high levels of immunosuppression are required, increas-
ing morbidity and mortality.
Tacrolimus-based regimens are the leading induction agents for
immunosuppressive therapies.
Table 2. Intestinal failure organ-specific complications.
Gastrointestinal Hepatobiliary Metabolic Renal
(1) Diarrhea
(2) Steatorrhea
(3) Gastric hypersecretion
(4) Small bowel bacterial overgrowth
(5) Changes in colonic flora
(1) Cholelithiasis
(2) Intestinal failure associated liver disease (IFALD)
(1) Malnutrition
(2) Micronutrient deficiency
(3) Fluid and electrolyte abnormalities
(4) D-Lactic acidosis
(5) Osteoporosis and osteomalacia
(1) Nephrolithiasis
(2) Chronic renal failure
Table 1. Different etiologies of short bowel syndrome.
Adults Children Infants
(1) Postoperative complications
(2) Inflammatory bowel disease (IBD)
(3) Ischemic bowel/mesenteric vascular disease
(4) Trauma
(5) Irradiation
(6) Neoplasia
(7) Other benign causes
(1) Postoperative complications
(2) Trauma
(3) Neoplasia
(4) Neuropathic/myopathic disorders
(5) Pseudo-obstruction syndrome
(1) Necrotizing enterocolitis (NEC)
(2) Bowel atresia
(3) Abdominal wall defects/gastroschisis
(4) Midgut volvolus
(5) Neuropathic/myopathic disorders
678 N. ZORZETTI ET AL.
QOL and intestinal function while weaning off HPN, decreas-
ing both the risk of IF and complications, it is important to
entrust patients to expert teams in Intestinal Rehabilitation
Centers: these expert teams can also list for ITx with the correct
time and appropriate type of transplantation [19].
In recent years, different molecules (hormones, proteins,
and growth factors) have been identified as steps in the IA
after an MSBR. Their research for the diagnosis and treatment
of SBS has raised considerable interest in the so-called ‘pro-
teomic therapy’ [6]: the raising interest for these substances is
justified by the fact that they can promote, after intestinal
resections, crypt and villus enterocytes expansion, increasing
absorptive function in order to obtain enteral independence.
Epidermal growth factor (EGF), glucagon-like peptide-1 (GLP-1),
and glucagon-like peptide-2 (GLP-2) are the molecules that
seem to regulate gene expression, during proteomic analyses
conducted subsequently to MSBR, and could have a role in the
clinical therapeutic field [6].
Among these, glucagon-like peptide-2 analogs (teduglutide
also known as GATTEX®, glepaglutide also known as ZP 1848
developed by Zealand Pharma, and apraglutide also known as
FE 203,799 developed by VectivBio) have been crucial and
allowed for dramatic clinical progress in the treatment of
SBS: many studies have been conducted on the subcutaneous
administration of Human GLP-2 (hGLP-2) and/or its synthetic
analogous, teduglutide, resolving in improvement of intestinal
absorption and creatinine clearance, reduction in fecal weight,
and improved maintenance of fluid and electrolytes, diminish-
ing the need and dependence for HPN [28–31].
In the United States teduglutide was approved by FDA for
adults in 2012 [32] and subsequently for children over the age
of one year in 2019 [33]. The other molecules are still in the
experimental stage as more research will focus on the safety
and long-term side effects. Also defining the correct timing to
initiate these therapies is crucial such as, it is, to underline that
the high cost of these drugs may limit their future clinical
application.
4. Non-transplant surgical techniques and
autologous gastro-intestinal reconstruction
The first surgical treatment of SBS is prevention preserving,
during surgical procedures, as much bowel as possible and,
whenever feasible, the ileocecal region; if intestinal resection is
necessary, the anatomy and the length of the remaining
bowel must be recorded in detail to guide the subsequent
approach [34]. However, even in this case, the evolution in the
management of chronic SBS in the last decade has made
transplantation rarely necessary.
If complications of SBS occur, nontransplant surgical tech-
niques are frequently used for autologous gastrointestinal
reconstruction (AGIR) to improve intestine function and
reduce HPN dependence and its complications, trying to
avoid ITx.
The most recent analysis of the prevalence of HPN noted
that the most common indication in adults was SBS (24%),
above all through a PICC line [35]. Although the catheter is the
lifeline of these patients, that are correctly educated regarding
proper care, complications can occur such as thrombosis,
central line associated bloodstream infection (CLABSI), or
damage; management of these complications include respec-
tively anticoagulation, IV antibiotics ± removal of CVC and
adequate repairs thanks to specific kits [35]. A recent new
tool is Taurolidine, a derivative of taurine, that prevents micro-
bial adhesion to catheter surfaces and biofilm formation, redu-
cing CLABSI when compared with eparine group but larger
trials are needed [36].
Other common HPN complications include liver failure-
induced liver injury (IFALD) and hyperglycemia. For what
that concern IFALD, prevention is the real goal limiting hepa-
totoxic agents with cycles of HPN and avoiding overfeeding
[37]; for the second one therapy with insulin, added directly to
HPN or provided subcutaneously, is indicated [35].
The same factors concern pediatric population [38].
So, the main goal to reach in SBS patients, both adults and
in children, remains the increased enteral tolerance and wean-
ing from PN support; when the achievement of enteral auton-
omy is not reached, the inability to progress in enteral
tolerance exposes these patients to long-term complications
of PN so, increasing morbidity and mortality: in this context,
with a personalized and multidisciplinary approach, cautiously
selected AGIR can facilitate the fulfillment of enteral auton-
omy, maximizing the absorptive potential of the remaining
gut [39]. When AGIR is not possible or not successful, ITx is
a valid option, alone or in combination with other organ
transplants [21].
These surgical techniques produce excellent results in
terms of enteral autonomy when associated with Intestinal
Rehabilitation Programs (IRPs) and represent, especially in
Western countries, a new frontier for the management of
SBS as they allow specialists to combine their knowledge
and better deal with these complex cases [40].
This approach stemmed from the management of pediatric
patients, combined with the caregivers’ support to patients
and families, increased survival rate in >70% of IRPs centers
[41–43].
Enteral autonomy can be achieved in approximately 25% of
patients subjected to AGIR, reaching 30% of patients when
intestinal segments are dilated [44].
The main targets of autologous intestinal reconstructive
surgery fall into two main surgical settings [39, Box 1]; some
procedures, each with its tailored indications and clinical
applications, are designed to achieve one of these goals
while others can act above multiple aspects.
The most common techniques of autologous intestinal
reconstructive surgery are:
LILT: designed to double the length of dilated SB loop
(≥4 cm) [45];
STEP : this lengthening procedure is quite ‘revolutionary’
as it avoids intestinal anastomosis (and its surgical
Box 1. Targets of autologous intestinal reconstructive surgery
1) Slow down intestinal transit increasing contact time between intestinal
mucosa and nutrients;
2) Reduce small bowel dilatation decreasing stasis and bacterial overgrowth
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 679
complications); it can be performed after LILT if re-
dilatation occurs and can also be performed on the
duodenum [45]. Clinical applications are widely
described in literature [46–48]. Also repeated STEP (Re-
STEP) is a possibility to further lengthen the SB when
enteral autonomy is not achieved [48–50];
SILT: is the newest lengthening technique. It has the
purpose of solving STEP’s main issue: the change of
intestinal muscle fibers’ orientation after stapling.
Limited clinical applications have been reported in litera-
ture so far [51];
Reconnection surgery without autologous gastrointestinal
reconstruction: it has long been used. The presence of the
colon in continuity with the shorter small bowel could
improve the recovery from SBS after medical rehabilita-
tion [52]. The procedure is required when there is no
small bowel dilatation. Only recently [25] it has been
suggested in an adult population that a residual small
bowel length of a minimum of 75 cm, even if recon-
nected to a part of the colon without autologous gastro-
intestinal reconstruction, seems able to produce an HPN
independence after a minimum period of 1 year-
intestinal rehabilitation.
In conclusion, STEP is the preferred procedure, probably
because it is the easiest technique: there is no need for
bowel suture and there is less chance of damaging the mesen-
tery vascularization [53].
Although AGIR achieves better long-term survival, ITx
shows better reestablishment of nutritional autonomy but,
on the other side, ITx has major disadvantages, complications,
and higher costs when compared to AGIR. In cases where
complications of PN occur, the patient cannot be weaned off
PN and the bowel is too short, the patient should be referred
to a transplant center to be evaluated for transplant treatment
as soon as possible because the prognosis after ITx worsen
when performed in patients with poor conditions and/or liver
failure. Nevertheless, it is still a valid option in patients with
organ failure, and a surgical algorithm has been developed to
select patients for AGIR or transplantation, to be used in
relation to the organ failure of the patient [45].
5. Small bowel transplant: changing indications and
recent progress
5.1. Indications and criteria for transplant
If a patient has permanent IF he has to be managed by
dedicated multidisciplinary intestinal rehabilitation team [21]
in order to evaluate the possibility of an ITx when PN fails [54–
56] Table 3 or if there are no alternative surgical techniques
(unreconstructable GI tract).
According to the American Society of Transplantation,
more indications have been added to the failure of PN
because of the high risk of death attributable to underlying
diseases [Box 2a].
Furthermore, subsequent experts’ indications for ITx [57]
have been annexed, considering both the high risk of death
and the high morbidity or low acceptance of PN [Box 2b].
In the pediatric population, a subset of children with intest-
inal failure remaining dependent on PN will develop life-
threatening complications arising from therapy. Since 2001,
ITx is recommended for this selected group affected by life-
threatening complications, warranting consideration of intest-
inal ITx, similar to the adult population. Children with life-
threatening complications of IF and PN therapy must be
early identified, that is in time to receive suitable donor organs
before they become critically ill [58].
According to the latest analysis confirmed by a recent mul-
ticenter study [58], the pediatric listing criteria for ITx should
be revised to reflect the changes in the natural history of
pediatric IF that have occurred as a result of modern therapies.
In addition, this analysis would suggest that it is possible to
predict who are the children with a high risk of death and the
need of an ITx with a 98% probability, given they meet two
out of three of the proposed criteria [Box 3].
In Europe, the new guidelines agree with the American
ones considering only the actual risk of death on HPN
[54,55]. Different data-reports underline also the fundamental
role of the Intestinal Rehabilitation Center for patients with
chronic intestinal failure (CIF), according to previous docu-
ments and recommendations [59,60], (Table 4).
Particular attention must be paid nowadays to ‘re-
transplantation’ that is a new reality and a current indication
if the failure of the first graft occurs, representing up to 8–9%
of transplant activity worldwide [55]; if listed for retransplant,
most were listed for a liver-containing allograft [61].
In conclusions, a lifesaving ITx is a mandatory therapeutic
strategy for patients that follow the previous indications; for
Box 2a. Extended indications for ITx (part 1)
Recurrent catheter-associated bloodstream infection (CABSI), especially
fungal
Unreconstructable GI tract
Complete porto-mesenteric thrombosis
Tumors involving the hepatic hilum or root of the mesentery
Frozen abdomen
Box 2b. Extended indications for ITx (part 2)
1) High risk of death attributable to the underlying disease
Desmoid tumors associated with familial adenomatous polyposis;
Congenital mucosal disorders (eg, microvillus atrophy and intestinal
epithelial dysplasia);
Ultrashort bowel syndrome (gastrostomy, duodenostomy, residual small
bowel 10 cm in infants and 20 cm in adults).
2) IF with high morbidity or low acceptance of PN
IF with high morbidity (frequent hospitalization, narcotic dependency) or
inability to function (eg, pseudo-obstruction disease, high-output stoma);
Patient’s unwillingness to accept long-term PN (eg, young patients)
Box 3. Current era listing criteria for pediatric ITx.
2 admissions to Intensive Care Unit (ICU)
Loss of 3 central venous catheter (CVC)sites
Persistent elevation of conjugated bilirubin ( 75 µmol/L) following 6 weeks
of lipid strategies
680 N. ZORZETTI ET AL.
those who do not have a life-threatening condition,
a preemptive ITx, in the future and in selected cases, might
be considered an indication itself [62].
Taking into account all the recommendations, it is impor-
tant to avoid any delayed referral of these patients to
Intestinal Rehabilitation Centers or directly to a transplant
center in order to choose the correct listing time and graft
type for ITx, bettering the proper allocation of the organs [55].
All unsuccessful prolonged attempts at intestinal rehabilitation
must be avoided because of their consequences, which repre-
sent a risk for early survival, such as the formation of anti-HLA
antibodies or multi-drug-resistance, hazarding ITx if eventually
performed [63–65].
Regarding the waiting list considerations, there has been
a change in trend over time. Initially, most candidates were
on the waiting list for less than 90 days, while over the last
decade more than 50% of the listed patients wait for over 1
year, and 36% over 2 years, making the mortality risk in the
list very high [60]: new indications increase the demand for
suitable donor intestines [62,66,67]. Because of the demand
for so-called ‘excellent donors’ and the related strict criteria,
donations are very rare and the waiting time could be very
long. Lastly, even if the number of new patients on the
2020 waitlist increased over the previous year, with an
increase of adult candidates rather than <18 years, the
trend over the last decade has been marked toward
a reduction of listing.
The overall pretransplant mortality for ITx candidates was
6.5 deaths per 100 waitlist years in 2020 and rates varied by
age, in general, were higher for adults (15.1 deaths per 100
waitlist-years vs 1.9 deaths per 100 waitlist-years in the
pediatric population) and it was inferior for isolated ITx
when comparing the intestine-liver combined trans-
plant [61].
5.2. Surgical techniques
ITx can be isolated or combined with other organs depending
on patient anatomy and comorbidities [55,68], including
a non-vascularized or vascularized abdominal wall [69] when
necessary.
High-volume centers of ITx (>5 patients per year for 5 years)
report for adult patients an overall 5-year survival with
a functioning graft of less than 70%, which is inferior to the
5-year survival obtained on HPN; in the pediatric population,
there is a group of infants and children who have successfully
ended PN and are unlikely to have experienced the morbid-
ities that warrant consideration for transplantation [70,71].
The main graft options are isolated intestine, liver intes-
tine and multi-visceral transplant; in the adult recipient, the
most common grafts used are isolated intestines, when liver
function is preserved, and multi-visceral grafts [68].
Proper graft selection for each recipient represents the
basis of surgical success. Donors’ criteria that have to be
evaluated are the gastrointestinal history, the absence of any
evidence of intestine trauma, hemodynamic stability, graft
size, and immunologic compatibility. Moreover,
a determinant component of any type of intestinal graft is
the jejunoileum [68].
Table 3. Accepted indications for ITx.
(1) Failed TPN:
liver failure-induced liver injury (IFALD);
thrombosis of the major central venous channels (when there is the involvement of two or more major ventral veins, there is a life-threatening complication and
failure of TPN therapy);
frequent-line infection and sepsis, requiring hospitalization;
multiple episodes of severe dehydration (diarrhea) despite intravenous fluid supplement
(2) Approved Transplant Facilities:
ITx is covered by Medicare if performed in an approved facility (10 intestinal transplants per year with a 1-year actuarial survival of 65% using the Kaplan–Meier
technique).
Table 4. Revised criteria related to wait-list for ITx.
Evidence of advanced or progressive IFALD with:
a. persistent hyperbilirubinemia >75 mmol/l (4.5 mg/dl) > 2 months;
b. any combination of elevated serum bilirubin ± sub-normal albumin or elevated INR ± portal hypertension and hypersplenism > 1 month (without associated
infections).
Notes:
the stage of liver disease is relevant for both the timing of transplantation and the type of transplant (isolated ITx or combined liver-ITx)
liver biopsy remains the gold standard
Thrombosis of three out of four upper body central veins (left subclavian and internal jugular, right subclavian and internal jugular) or occlusion of
a brachiocephalic vein in children (while in adults this criterion should be evaluated on a case-by-case basis).
Life-threatening morbidity in the setting of indefinite PN dependence of either anatomical or functional cause, as suggested by:
(a) In children after two admissions to an Intensive Care Unit
(b) In adults, on a case-by-case basis taking into account catheter-related bacteremia/sepsis (CRBS) ± of severe dehydration and/or electrolyte disturbances
Invasive intra-abdominal desmoids in adolescents and adults (mostly as a sequela of familial adenomatous polyposis) above all when greater than 10 cm
with immediate listing
Acute diffuse intestinal infarction with hepatic failure (sometimes needing a multivisceral transplantation).
Failure of first intestinal transplant (considering that re-transplant is a possibility when choosing the correct timing).
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 681
It is important to underline that among the pediatric and
adult populations the ‘liver intestine’ graft differs. In pedia-
trics, it could be used to indicate a composite liver-pancreas-
intestine graft listing the candidate for all three organs; the
surgical technique is easier thanks to the foregut preservation
and en-bloc transplant creating a portocaval shunt [72]. In the
adult population often, there is a combined liver-intestine
graft: the liver is transplanted first, preserving the foregut
anatomy, it is then followed by an isolated ITx: this is called
‘non-composite’ graft [73], requiring an intact and patent
portal vein. A hepatic biopsy is the gold standard, over endo-
scopic ultrasound and transient elastography, to assess the
entity of liver disease severity and the need for simultaneous
intestine liver transplant [74–76].
In some cases, there is the indication for multi-visceral
transplantation (MvTX), which is a complex procedure that
classically includes stomach, pancreaticoduodenal complex,
small bowel, and spleen with (MVTx) or without (modified
multi-visceral transplantation, MMVTx) the liver; it requires
also the replacement of all abdominal organs with an en-
bloc graft [77]. The main indications are multiple previous
surgeries, porto-mesenteric thrombosis, and abdominal
tumors above all desmoids. Splanchnic thrombosis is
a challenging condition but MvTX manages to reestablish
the venous anatomy and physiology of the abdominal cavity,
resolving completely the effects of portal hypertension and
the baseline disease [78]. Complete foregut resection followed
by MvTX could be a therapeutic option for large intra-
abdominal tumors (desmoid, well-differentiated neuroendo-
crine and gastrointestinal stromal ones) that are slow growing
and tend to locally invade vessels and viscera [79].
Furthermore, the inclusion of a colon component in con-
tinuity with the jejunoileum can be added to all these grafts,
both in pediatric patients and adults [68]: this option leads to
a series of advantages such as 6% superior graft survival at 3
years, higher frequency of formed stool and significant enteral
independence improving QOL [80].
A recent paper [81] has analyzed the outcome following
multi-visceral transplantation without a stoma, routinary cre-
ated during ITx to allow follow-up graft endoscopy and
biopsy. It seems a feasible model without a difference in
clinical outcomes with also better fluid balance, abdominal
wound care, and patient satisfaction. A longer follow-up with
more cases of this potentially attractive procedure will be
needed.
Common surgical complications following ITx are intestinal
obstruction, anastomotic stricture, anastomotic leak, bowel
perforation, enterocutaneous fistula, allograft volvulus, and
bowel internal hernia; they can result in allograft ischemia
and necrosis and may contribute to both early and late graft
loss [82].
5.3. Immunosuppressive therapy
The great amount of lymphoid mucosal tissue, present in the
small intestine and very well represented in ITx recipients with
the subsequent immune response is the primary cause of graft
loss and rejection.
There has been evidence of improvement in graft and
patient survival rates only up to 1-year; beyond that limit
data is unclear [83]. In order to achieve long-term graft survi-
val, it is necessary to obtain a valid immunosuppression (IS),
reduction to the minimum of IS side-effects, early diagnosis of
acute cellular rejection (ACR), accurate ACR treatments, and
improvement of allograft tolerance.
A fundamental part of ITx immunosuppressive therapy is
represented by the induction made via classical and emer-
ging protocols. Antithymocyte globulin (Thymoglobulin), that
induces T-cell depletion, represents the mainstay of IS
induction and is usually used as a primary induction
agent [84]. Basiliximab, a monoclonal antibody that targets
the interleukin-IL2 receptor, is used above all for unsensi-
tized ITx recipients promoting the engraftment of regula-
tory T cells, decreasing steroids and global T cell-depleting
agents [85,86]. Alemtuzumab, a monoclonal antibody that
targets CD52 on mature T cells, leads to excellent 1-year
survival with low rejection rates in combination with tacro-
limus [87]. The emerging protocols include new strategies
such as the Leuven protocol, which is based on broad-based
IS, that promotes a regulatory environment, utilizes donor-
specific blood transfusion, avoids high-dose steroids and
calcineurin inhibitors, and minimizes ischemia-reperfusion
injuries and endotoxin translocation [86]. Moreover,
delayed donor bone marrow infusion combined with ITx
represents a strategic trial of tolerance induction [87,88].
Due to the lack of literature about major induction thera-
pies, the current consensus is based on a comparison of large
single-center series. The safest and most established agent is
Thymoglobulin, even if there is a risk of infective complica-
tions. For unsensitized patients, Basiliximab is a good option
to prevent rejection and lastly Alemtuzumab may reduce both
rejection and GVHD: the first monoclonal antibody is reported
to avoid adverse effects while the use of the second one has
not been clearly evaluated to assess the malignant or infective
risks [89].
Maintenance therapy is more standardized: tacrolimus-
based regimens are the mainstay, used in 73% of recipients,
combined with corticosteroids (37.4%) [61], while mycophe-
nolate mofetil and sirolimus (mammalian the target of rapa-
mycin – mTOR inhibitor) are used in fewer protocols [90,91]. In
addition to standard therapy, tapered off by 6 months post-
transplant, the polyclonal antibody rabbit anti-thymocyte glo-
bulin [92] combined with the chimeric anti-CD20 monoclonal
antibody Rituximab has been used as dual-induction therapy
at Miami Transplant Institute since 2013, based on previously
reported successful results [93]; anti-thymocyte globulin may
also protect against reperfusion injury [94–96]. According to
a recent report [97], a significant reduction of ACR risk can be
obtained when Rituximab is added to the induction therapy,
making it more efficient in removing B cells and therefore
reducing the risk of developing DSAs against the intestinal
graft.
To better understand and avoid rejection, past studies have
investigated the role of antibody-mediated rejection (AMR)
and donor-specific antibodies (DSAs) in ITx: DSAs can be
found prior to transplant or can develop de novo after the
procedure. They can persist after IS doubling the risk of acute
682 N. ZORZETTI ET AL.
humoral rejection and increasing the risk of chronic rejection
[98]. It seems that a combined transplantation with a liver
graft clears the preformed DSAs with a protective role [99].
Moreover, rejection can be induced also by preexisting or de
novo non-HLA antibodies that could act as triggers to a DSA-
mediated allogenic response [100].
Desensitization is a potential option to reduce the number
of preformed antibodies, acting on human leukocyte antigen
(HLA) thus reducing the risk of early graft loss prior to alloca-
tion. Nevertheless, its effective application is limited [101].
Apart from plasma exchange and high-dose intravenous
immunoglobulin (IVIG), emerging strategies to treat it have
been found such as rituximab (a monoclonal antibody against
CD20 found on B-cells) and bortezomib (a selective inhibitor
of the 26S proteasome and nuclear factor-kB that has anti-
plasma cell activity) [102]. However, there is still a lack of an
optimal algorithm able to use immunologic biomarkers or of
the identification of specific patient characteristics to guide its
clinical application.
Because of the immunological complexity of the intestine,
compared with other solid organs, rejection and graft-versus-
host disease (GVHD) frequently occur, requiring high levels of
IS thus increasing morbidity. Standardized and effective ther-
apeutic protocols are still lacking in literature due to the
paucity of centers with experience and the overall low volume
of ITx [89]. Recent insights into specific immunological path-
ways have led to novel and targeted immunomodulatory
strategies that aim to improve outcomes [103].
Classical therapies to treat allo-rejection include high-dose
steroids, increasing calcineurin (Tacrolimus), or mTOR inhibitor
levels [89]. The combination of thymoglobulin with steroids
and rituximab seems to reduce rates of chronic rejection and
PTLD [94] and is reserved for severe or steroid refractory
rejection, but increases the risk of opportunistic infections
[104]. Basiliximab induction therapy reduces the incidence of
ACR after ITx without increasing the incidence of infectious
complications. The use of Alemtuzumab as antirejection ther-
apy seems to be associated with higher rates of PTLD [105]
and infection in some series. Although it is occasionally possi-
ble to treat successfully refractory rejection with strategies
targeting antibodies (IVIG, plasmapheresis) or B cells (rituxi-
mab), it is necessary to find better strategies [106]. Emerging
agents to treat allo-rejection are: 1) vedolizumab, recently
reported off-label and used successfully for the treatment of
ACR after ITx [107]; 2) infliximab, recently described for the
suppression of inflammation [108,109]; 3) bortezomib, which
seems to show effectiveness [110]; and 4) eculizumab,
reported as successful by a single paper [111]. New strategies
under investigation in human intestinal rejection are repre-
sented by the use of probiotics and/or fecal microbiota trans-
plant [112,113].
In terms of immune-monitoring, a recent paper [114] ana-
lyzes peripheral blood samples and intestinal allograft biopsies
from ITx patients with severe rejection. It concludes that it
would be advisable to keep monitoring both parameters in
allograft patients.
Strong IS induction agents such as alemtuzumab and anti-
thymocyte globulin are administered when severe rejection
occurs. When this treatment fails, the re-transplant is a valid
option. Nevertheless, even though the overall short-term sur-
gical outcome has improved in the last decades, the need for
a second round of IS, in patients undergoing re-
transplantation, leads to a state of severe immune deficiency,
together with bone marrow suppression, thrombocytopenia,
and leukopenia thus resulting in infections, malignancies and
an increase in morbidity and mortality [115]. According to this,
to improve long-term results and avoid super-immunization,
prolonged immunosuppression-free periods prior to re-
transplantation should be recommended, in order to reconsti-
tute the host’s immunity. Moreover, it is advisable to use
individualized IS strategies guided by patients’ immunocom-
petency [108].
Recently, there have been reports of operational tolerance
after ITx without evidence of rejection in the absence of
immunosuppression for at least one year [116].
Post-intestine transplant infections (bacterial, viral, and
fungal) are ubiquitous, especially in the pediatric popula-
tion, with rates of 90%, 25%, and 75%, respectively. The
infectious risk was not greatly impacted neither by the
inclusion of the liver or colon component nor by adding
rituximab to the induction protocol [117]. Therefore, in
patients with a lower risk of graft rejection, a lower IS
induction is recommended.
5.4. Outcomes
According to recent data, the five-year survival rate for ITx is
almost 50.5%, unlike that of the liver (72.8%) or kidney
(78.5%). These numbers are due to high rates of rejection,
transplant complications, and related treatment. As
a consequence, ITx is still commonly used as a salvage therapy
for TPN failure or life-threatening complications rather than
a real chance for long-term success [118].
Comparing data of the last decade, a trend of improvement
in 6- and 12-month isolated ITx graft survival: for transplants in
2019, only 4 3.3% of grafts had failed by 6 months and 6.7% by
1 year compared to 23.8% and 31.7% of transplants in 2009
without, on the other side, improvements in 3- and 5-year
(around 40% and 50% respectively) [61].
Among intestine-liver transplant recipients, 42.1% had graft
failure at 6 months and 47.4% at 1 year in 2019, 55.2% at 3
years in 2017, 69.0% at 5 years in 2015, and 75% at 10 years in
2010; there has been no consistent improvement in graft
survival over the last 10 years for liver-inclusive allografts [61].
Comparing ITx with or without a liver in 2013–2015, 1- and
5-year graft survival was 72.8% and 44.8%, respectively, for
recipients 18 years or older and 76.0% and 61.3% for those
younger than 18 years old. One- and 5-year graft survival was
77.7% and 48.9%, respectively, for intestine recipients and
70.6% and 53.2%, respectively, for intestine-liver recipients
[61]. Patient survival at 1 year for transplants in 2013–2015
was higher for recipients of an intestine alone (83.7% vs
75.6% for intestine-liver); however, by 5 years, the patient
survival was identical, at 58.7% for both allograft types [61].
The number of recipients with a functioning ITx reached
a plateau at 1,217, on 30 June 2020, excluding isolated reci-
pients younger than 18 years old. This is probably due to the
fact that ACR and its clinical consequences, that remain the
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 683
leading cause of allograft loss and mortality, not considering
the new strategies of IS [85,104], varies by age and transplant
type in the first year after ITx [61].
Among recipients in 2018–2019, incidence of acute rejec-
tion was highest in adult intestine recipients (41.2%) and low-
est in adult intestine-liver recipients (35.4%) [61].
The most common post-ITx complications are:
postoperative infection: reported in up to 97% of recipi-
ents, it is the most frequent complication and cause of
graft loss overall due to over-IS and bacterial transloca-
tion [83];
post-transplant lymphoproliferative disease (PTLD): charac-
terized by uncontrolled B-cell proliferation, often due to
the de-latentisation of Epstein–Barr virus infection fol-
lowing immunosuppression [68]. It can be treated with
a reduction in IS to bolster T-cell responses or, secondly,
with rituximab [119]. Pediatric patients are subject to
a higher risk [120];
graft-versus-host disease (GVHD): compared to other solid
organ transplantation, GVHD is more common after ITx
and usually appears early but it can be delayed beyond
the first-year post-transplant. It tends to affect the skin,
native bowel, and native liver, manifesting as a rash,
enteritis/colitis, and hepatitis, respectively [120] rather
than the allograft itself. The clinical evaluation is the
basis for the initial diagnosis supported, when necessary,
by skin biopsy. Steroids remain the first line of therapy
for both acute and chronic GVHD [121]. According to
a recent single-center study, accurate donor selection
and adequate IS may reduce GVHD incidence improving
survival. In the pediatric population, younger donors
seem to reduce GVHD risk, whereas including the colon
with the ITx and/or repeat transplantation seems to
increase it. Lastly, in the adult population, the use of
Interleukin-2 plus sirolimus after ITx reduces GVHD risk
[122]. Alemtuzumab shows a significantly favorable
induction effect reducing the development of post-
transplant GVHD, acting both on graft-versus-host and
on host-versus-graft responses [123];
chronic rejection: it has been recognized as one of the main
causes of late graft loss. It is particularly common in isolated
small bowel transplantation and has a greater risk in teen-
agers and young adults as there is a more frequent non-
adherence to immunosuppressive medications [124].
Clinically, it manifests as a progressive and irreversible loss
of intestinal allograft function over a period of 3 months in
the absence of other causes, it is unresponsive to pharma-
cologic treatment and requires the re-institution of PN sup-
port [125]. In case of sepsis or severe rejection, removal of
the graft and IS can be lifesaving and re-transplantation can
be achieved once the patient recovers [126];
ischemia-reperfusion injury: it represents a central topic of
recent research that aims to find novel approaches, such
as luminal interventions during cold storage that could
prolong the storage time and alleviate ischemia-
reperfusion injury. It must be taken into consideration
that ischemic susceptibility differs among species
[127,128].
Rejection and infectious complications remain the most com-
mon causes of morbidity and mortality; it is, therefore essen-
tial to monitor closely the intestinal graft to prevent such
occurrences. The absence of a reliable biochemical marker to
help in the graft status follow-up and to promptly diagnose
any dysfunction related to ACR or primary disease recurrence
has led to the need for frequent surveillance via endoscopy
and biopsy (E&B) [129–131]; according to this condition, the
creation of a stoma during ITx can be considered a routinary
act [81], impacting the QOL of the patient.
Likewise, surveillance represents a cornerstone in follow-up
after ITx but the current lack of evidence-based criteria to
create a diagnostic algorithm renders endoscopy and biopsies
necessary [132]. A recent report underlines that the natural
history of disease recurrence after gut failure has yet to be
fully defined but the absence of a universal IS management
protocol is a relevant factor [133].
Long-term patients and graft survival outcomes after ITx
have remained somewhat static in recent years but experi-
enced centers are now reporting improved long- and very
long-term data. Hope for further improvement exists, as stra-
tegies to promote protective T-regulatory cell mechanisms are
developed and the understanding of humoral immunity
increases. Nutritional and QOL outcomes are generally good
and may be better in the future than the ones in patients on
home PN. The psychosocial impact of ITx is high on recipients
and families but most patients reintegrate into society living
fulfilling and productive lives [134].
There is a need for reliable noninvasive diagnostic means in
identifying ITx rejection. A recent paper took into considera-
tion the use of EUS (endoscopic ultrasound) as a tool for
assessing potential CR in long-term follow-ups of intestinal
allograft recipients. Nevertheless, EUS and enteroscopy, while
providing detailed information on the intestinal graft mor-
phology and rheology, are still associated with biopsies mak-
ing partially invasive techniques [135]. A completely
noninvasive method is peripheral blood monitoring using
different potential molecules: plasma citrulline concentration
(CIT), well-known for its role as a noninvasive marker of ACR in
ITx recipients [136], and more recently REG3α, an antimicrobial
peptide secreted by intestinal enterocytes and Paneth cells.
The latter could become a screening biomarker as it can
predict acute allograft rejection a week earlier than the evi-
dence of histopathological changes [137]. Lastly, immunophe-
notyping of peripheral blood T cells has demonstrated its
efficacy in better understanding the immune-mechanisms of
allograft dysfunction, in identifying potential biomarkers in ITx
and in selecting IS therapies [138].
Nevertheless, further studies are needed with larger
cohorts to validate these noninvasive diagnostic tools.
6. OPTN data on rates and trends
According to recent studies, in 2020 ITx donors in the U.S.A
were mainly represented by head trauma or anoxia patients,
usually younger than 18 years [61].
Globally, during the last decades, the trend of ITx is declin-
ing (apart from a small rise in 2019) and adults consistently
exceed the pediatric reports. The pediatric waitlist mortality is
684 N. ZORZETTI ET AL.
decreasing whilst it keeps being relatively stable for adults
[61]; the same occurs for the number of centers performing
this procedure and the annual program volume (median 3.5
ITx in 2020). Although there has not been any apparent
improvement in post-ITx graft survival in the years following
the first, it seems that these data will stabilize while the annual
number of ITx will continue to decline [81].
7. Post-transplant quality of life
Different questionnaires exist in literature for adults and chil-
dren to evaluate the quality of life (QOL) in patients affected
by IF. In both IF groups, QOL is considered generally good.
With regard to ITx, patients assume to have a QOL almost
equal to the general population but the results are completely
different when considering parents/caregivers that report,
both for HPN and ITx, a lower QOL suggesting a negative
perception on family activities. In conclusion, ITx has a good
QOL; however, the possible complications induced by IS or
rejection did not establish the indications for preemptive
transplantation based on QOL [139].
In the future, through controlled studies, specific QOL tools
(such as the combination of HPN-QOL and EuroQol-5) could
provide the guidance needed for ITx [62,140].
8. Cost-effectiveness of intestinal transplant
Cost should never be a prohibitive parameter in considering or
receiving an ITx or other surgical and non-surgical therapies
but, from a socioeconomic perspective, the healthcare burden
of irreversible IF is enormous. Although few studies discuss the
costs of HPN and ITx, no study has definitively established the
cost-effectiveness of ITx: extensive and exact data on costs are
lacking, but according to the literature ITx may become cost-
effective after 1–2 years in pediatric recipients who retain
their functional grafts [141].
The non-transplant surgical techniques are useful in
selected patients and have fewer advantages, including
costs, when compared to ITx [45].
Lastly, the high cost of teduglutide prohibits its widespread
use: special attention should be paid to how it is used, and
priority should be given at evaluating strategies to lower its
cost per patient [142].
9. Conclusions
In conclusion, over the last decade, wait-list mortality and
overall ITx rates are decreasing, especially in the pediatric
population [61] while remaining almost similar for adults.
The type of graft that is required is highly individualized
according to the patient’s original diagnosis and status. The
continuous improvement of 1-year graft survival is instead
unaccompanied by the same results in 3- and 5-year. At 5
year, there are no differences between survival rates of iso-
lated and liver-inclusive ITx but still lower than other solid
organ transplants.
It is important to refer to transplant centers selected
patients before they become too ill to be transplanted,
which of course plays a role in a poorer outcome compared
to other solid organ transplantations. One of the issues is that
although it is possible to prolong patient’s lives with IF, this
line of treatment leads to the accumulation of comorbidities
that translates into a more complex surgical procedure and
management.
The use of induction strategies, followed by tacrolimus-
based regimens, still remains the base of IS. The emergence
of both intestinal surgical and medical rehabilitation as speci-
fic subspecialties, due to the success of the larger transplanta-
tion centers, confirms that ITx is a life-sustaining treatment in
the critical case of IF, bettering QOL. Nonetheless, the trend
for the last decades is orientating toward less listing and
transplantation, notably in the pediatric population, consider-
ing, on one hand, the advances in IF care and, on the other
one, the absence of significant improvements beyond 1-year
graft survival. Currently, ITx in adults consistently exceeds the
pediatric population, and the number of listed adult candi-
dates is approaching one of the pediatric candidates [61].
Considering that overall ITx activity represents less than
0.5% of all organ transplants, a limited number of medical
professionals deals with this issue comprehensive of IF man-
agement [143]: it is mandatory, therefore, to assure adequate
education on this topic [144].
As the allocation of liver grafts remains a challenge, the
waitlist mortality persists for chronic IF patients requiring
a liver [145]. For such reason, isolated ITx should be sustained
even if, in these patients, chronic rejection occurs easily.
However, considering that re-transplantation is nowadays
a reality, it must be taken into account that over-IS easily
induces sepsis, which represents the main cause of post-
transplant death.
As there are only a few large centers that perform ITx
worldwide, they should unify efforts to address issues in
order to support those regions/nations without a program
and should cooperate performing multicenter studies to
advance more in this intricate field. Financial support to
a transplant recipient should include easier access to the
referral center for annual follow-up or if complications occur
[123]. The ultimate goal, for both the medical and surgical
transplant community, remains enteral autonomy benefiting
from rehabilitation, AGIR or ITx [123].
10. Expert commentary
Medical rehabilitation in SBS has dramatically improved with
the clinical introduction of Teduglutide while, on the other
hand, AGIR procedures gained success in lengthening and
tapering the remnant small bowel: preserving the ileocecal
valve represents the main predictive factor to achieve enteral
autonomy. Whereas the ileum has shown increased adaptive
function than the jejunum, also colon has great importance in
the digestive process: 1) in the adult population, a residual
small bowel length of a minimum of 75 cm, even if recon-
nected to a part of the colon, seems able to produce a TPN
independence; 2) a colonic graft is nowadays widely used
and 3) the terminal ileum is the selected segment for a living-
related donation.
The Intestinal Rehabilitation and Transplant Association
reached a consensus in 2019 on patients who have failed
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 685
medical and/or surgical rehabilitation. The latter should be
listed for ITx only if they are at actual risk of death on HPN
[Box 2a, 2b, 3].
Post-transplant long-term survival seems to have improved
if we consider the experienced US center (between 60% and
70% at 10 years), whilst it remains stable if we take into
account the International Transplant Registry (41% 10-year
survival). Main shared challenges still remain, such as: over-
IS, rejection, infection, and altered renal function. Chronic
rejection occurs with an incidence of 5–10% after intestinal/
multi-visceral transplant and allograft removal still represents
the main therapy because there is a lack of standardized
pharmacological strategies. Desensitization with
a conventional or novel technique could be adopted as
a protocol before ITx in those candidates who, thanks to
virtual crossmatching, have high levels of preformed donor
anti-HLA antibodies.
Using a de novo DSA monitoring plus treatment protocol
and efforts to establish formal histologic criteria for antibody-
mediated rejection, could further improve post-transplant
results. The adjunct of a liver graft, when re-transplanting,
could be helpful in a sensitized recipient due to its protective
effect against humoral immunity.
Technical surgical challenges for the closure of the abdo-
men at the end of ITx are represented by volume/edema of
donor graft and loss of abdominal domain in the recipient,
due to previous surgeries. Currently, new techniques based on
transplanting a full-thickness abdominal wall from the same
donor, vascularized or not-vascularized, have been adopted in
place of previous techniques such as graft reduction (espe-
cially liver and small intestine) or the volume increase in the
abdomen cavity.
Finally, nutritional outcomes are good with most recipients
achieving enteral autonomy with an unrestricted diet and QOL
improves over time in most psychological-emotional and
social areas. Pregnancy is possible after ITx although two
factors should be taken into consideration: the absorptive
function of the transplanted bowel and the higher need for
IS. Both of them have to be monitored closely with endosco-
pies and biopsies of the graft, to prevent episodes of rejection
and to preserve the fetus from temporary malnutrition.
Novel approaches such as manipulation of microbiota or
tissue bioengineering will be added to medical and surgical
rehabilitation in order to successfully treat short bowel syndrome.
Whenever unavoidable, the intestinal transplant should be
performed, but multiple causes of late graft loss including
chronic rejection, infection, graft-versus-host disease, post-
transplant lymphoproliferative disorder, and post-surgical
complications should be addressed and solved in the next
few years. Disease recurrence represents a drawback of the
procedure and should be faced in addition to neoplastic dis-
orders: disease recurrence could be suspected in patients with
pre-transplant Crohn’s disease, gut dysmotility, hypercoagul-
ability, metabolic syndrome, and some mucosal and neuro-
muscular disorders. Moreover, the role of ischemia-reperfusion
injury among causes of graft loss should be better established.
Graft failure after intestinal transplantation is often treated
by allograft removal due to a lack of effective drugs: multi-
visceral patients can also have the graft removed in refractory
rejection although they can be more challenging to manage.
Re-transplant of such a patient with another multi-visceral
graft is feasible but factors associated with anatomy, vascular
access, size, and sensitization should be evaluated before
making the decision of which organs have to be re-
transplanted.
While the importance of endoscopy and stool studies is
well known as the gold standard for monitoring, calprotectin
and citrulline plus measurements of immunoreactivity and
donor-specific antibodies represent promising tools in graft
follow-ups: for instance, the large amount of blood gran-
zyme-B or perforin before the clinical symptoms and endo-
scopic signs of acute cellular rejection, are produced by
activated recipient ‘circulating’ lymphocytes or by activated
donor-derived ‘resident’ lymphocytes? The question does not
represent a futile topic under the immune-surveillance point
of view. Nowadays, immunosuppression for intestinal trans-
plantation is mainly based on preconditioning protocols that
pre-treat a recipient, but earlier on the elimination by ex vivo
irradiation of mature resident lymphoid elements from the
bowel allografts was used to eliminate the GVHD risk, followed
by infusion of donor bone marrow cells in recipients to
improve tolerogenesis.
Preemptive transplantation in patients affected by irre-
versible intestinal failure is still far to be clinically applied. In
the future, it could be reserved for those patients identified
as the ones at high risk of complications and mortality
while on definitive long-term PN. A preemptive transplant
could be used as a rehabilitative procedure in a well-
selected population, among PN patients at risk of liver fail-
ure, repeated central-line infections, mesenteric infarction,
short bowel syndrome <50 cm or with end stoma, congeni-
tal mucosal disease, desmoid tumors: the development of
simulation models or pre-transplant scoring systems could
help in selecting the candidates to this preemptive
procedure.
Abbreviations
IF Intestinal failure
GI Gastrointestinal
PN Parenteral nutrition
SBS Short bowel syndrome
IA Intestinal adaptation
MSBR Massive small bowel resection
HPN Home parenteral nutrition
IFALD Intestinal failure associated liver disease
ITx Intestinal transplantation
SBL Small bowel length
ICV Ileocecal valve
QOL Quality of life
GF Growth factor
AGIR Autologous gastrointestinal reconstruction
IRPs Intestinal Rehabilitation Programs
LILT Longitudinal intestinal lengthening and tailoring
STEP Serial transverse enteroplasty
SILT Spiral intestinal lengthening and tailoring
IRTA Intestinal Rehabilitation and Transplant Association
ESPEN European Society for Clinical Nutrition and Metabolism
CRBS catheter-related bacteremia/sepsis
OPTN Organ Procurement and Transplantation Network
AMS Antibody-mediated rejection
DSAs Donor-specific antibodies
686 N. ZORZETTI ET AL.
IS Immunosuppression
ACR Acute cellular rejection
HLA Human leukocyte antigen
IVIG Intravenous immunoglobulin
GVHD Graft-versus-host disease
mTOR Mammalian target of rapamycin
E&B Endoscopy and biopsy
PTLD Post-transplant lymphoproliferative disease
MvTX Multi-visceral transplantation
MMvTX Modified multivisceral transplantation
CLABSI Central line – associated bloodstream infection
Acknowledgments
We wish to thank Ms Elena BALDISSONE for her invaluable support in
English editing.
Funding
This paper was not funded.
Declaration of interest
The authors have no relevant affiliations or financial involvement with any
organization or entity with a 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 or patents received or pending, or royalties.
Reviewer disclosures
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
References
Papers of special note have been highlighted as either of interest (•)
or of considerable interest (••) to readers.
1. Kappus M, Diamond S, Hurt RT, et al. Intestinal failure: new defini-
tion and clinical implications. Curr Gastroenterol Rep. 2016;18(9):48.
DOI:10.1007/s11894-016-0525-x
2. Pironi L, Konrad D, Brandt C, et al. Clinical classification of adult
patients with chronic intestinal failure due to benign disease: an
international multicenter cross-sectional survey. Clin Nutr. 2018;37
(2):728–738. DOI:10.1016/j.clnu.2017.04.013
3. Buchman AL. Intestinal failure and rehabilitation. Gastroenterol Clin
North Am. 2018;47(2):327–340. DOI:10.1016/j.gtc.2018.01.006
Excellent review on intestinal failure and rehabilitation by
a worldwide expert
4. Duggan CP, Jaksic T, Ingelfinger JR. Pediatric intestinal failure.
N Engl J Med. 2017;377(7):666–675.
5. Guillen B, Atherton NS. Short Bowel Syndrome. In: StatPearls [Internet].
Treasure Island (FL): StatPearls Publishing LLC; 2022:1. JanJul 18.
6. Zorzetti N, D’Andrea V, Lauro A. Proteomic biomarkers in short
bowel syndrome: are we ready to use them in clinical activity?
Expert Rev Proteomics. 2021;18(4):285–293.
7. Fjermestad H, Hvistendahl M, Jeppesen PB. Fasting and postpran-
dial plasma citrulline and the correlation to intestinal function
evaluated by 72-hour metabolic balance studies in short bowel
jejunostomy patients with intestinal failure. JPEN J Parenter
Enteral Nutr. 2018;42:418–426.
8. Lauro A, Lacaille F. Short bowel syndrome in children and adults:
from rehabilitation to transplantation. Expert Rev Gastroenterol
Hepatol. 2019;13(1):55–70.
9. McKee RF, Knight K, Leitch EF, et al. Changes in adult home
parenteral nutrition practice over 25 years. Clin Nutr ESPEN.
2021;45:170–176.
10. Barco S, Heuschen CB, Salman B, et al. Home parenteral
nutrition-associated thromboembolic and bleeding events: results
of a cohort study of 236 individuals. J Thromb Haemost. 2016;14
(7):1364–1373. DOI:10.1111/jth.13351
11. Gonzalez-Hernandez J, Daoud Y, Styers J, et al. Central venous
thrombosis in children with intestinal failure on long-term parent-
eral nutrition. J Pediatr Surg. 2016;51(5):790–793. DOI:10.1016/j.
jpedsurg.2016.02.024
12. Fonseca G, Burgermaster M, Larson E, et al. The relationship
between parenteral nutrition and central line-associated blood-
stream infections: 2009-2014. JPEN J Parenter Enteral Nutr.
2018;42(1):171–175. DOI:10.1177/0148607116688437
13. Gompelman M, Causevic E, Bleeker-Rovers CP, et al. Catheter-
related bloodstream infection management in patients receiving
home parenteral nutrition: an observational cohort study. Clin Nutr
ESPEN. 2022;50:155–161.
14. Fuglsang KA, Brandt CF, Jeppesen PB. Survival in patients initiating
home parenteral support due to nonmalignant short bowel syn-
drome compared with background population. Clin Nutr ESPEN.
2022;50:170–177.
15. Lezo A, D’Eusebio C, Riboldi L, et al. HPN standard of care and long-
term outcomes of CIF pediatric patients: twenty-eight years’ experi-
ence in a reference center. Front Nutr. 2022;9:868273.
16. Cloutier A, Bond A, Taylor MI, et al. Successful implementation of
remote video consultations for patients receiving home parenteral
nutrition in a national UK Centre. Frontline Gastroenterol. 2019;11
(4):280–284. DOI:10.1136/flgastro-2019-101257
17. Raphael BP, Schumann C, Garrity-Gentille S, et al. Virtual telemedi-
cine visits in pediatric home parenteral nutrition patients: a quality
improvement initiative. Telemed J E Health. 2019;25(1):60–65.
DOI:10.1089/tmj.2017.0298
18. Bering J, DiBaise JK. Home parenteral and enteral nutrition.
Nutrients. 2022;14(13):2558.
19. Adams SC, Gura KM, Seres DS, et al. Safe care transitions for
patients receiving parenteral nutrition. Nutr Clin Pract. 2022;37
(3):493–508. DOI:10.1002/ncp.10861
20. Choi YH, Kim MS, Kim CH, et al. Looking into the life of
technology-dependent children and their caregivers in Korea: lift-
ing the burden of too many responsibilities. BMC Pediatr. 2020;20
(1):486. DOI:10.1186/s12887-020-02388-z
21. Lauro A, Coletta R, Morabito A. Restoring gut physiology in short
bowel patients: from bench to clinical application of autologous
intestinal reconstructive procedures. Expert Rev Gastroenterol
Hepatol. 2019;13(8):785–796.
22. Pironi L. Definitions of intestinal failure and the short bowel
syndrome. Best Pract Res Clin Gastroenterol. 2016;30(2):173–185.
DOI:10.1016/j.bpg.2016.02.011
•• Outstanding review on intestinal failure and short bowel syn-
drome by a well-known worldwide expert
23. Fung JJ. William Hunter harridge lecture: the changing face of
short-gut syndrome management. Am J Surg. 2017;213(3):448–451.
24. Kong W, Wang J, Ying R, et al. A potential anatomic subtype of short
bowel syndrome: a matched case-control study. BMC Gastroenterol.
2016;16:12.
25. Lauro A, Cirocchi R, Cautero N, et al. Reconnection surgery in adult
post-operative short bowel syndrome < 100 cm: is colonic conti-
nuity sufficient to achieve enteral autonomy without autologous
gastrointestinal reconstruction? Report from a single center and
systematic review of literature. G Chir. 2017;38(4):163–175. DOI:10.
11138/gchir/2017.38.4.163
26. Sjoberg Bexelius T, Ahle M, Elfvin A, et al. Intestinal failure after
necrotising enterocolitis: incidence and risk factors in a Swedish
population-based longitudinal study. BMJ Paediatr Open. 2018;2
(1):000316. DOI:10.1136/bmjpo-2018-000316
27. Pederiva F, Sgrò A, Coletta R, et al. Outcomes in patients with short
bowel syndrome after autologous intestinal reconstruction: does
etiology matter? J Pediatr Surg. 2018;53(7):1345–1350. DOI:10.
1016/j.jpedsurg.2018.03.009
Very interesting paper correlating etiology to surgical recon-
struction on SBS by a leader in this field
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 687
28. Zhu C, Li Y. An updated overview of glucagon-like peptide-2
analog trophic therapy for short bowel syndrome in adults.
J Int Med Res. 2022;50(3):3000605221086145.
29. Suzuki R, Brown GA, Christopher JA, et al. Recent developments in
therapeutic peptides for the glucagon-like peptide 1 and 2
receptors. J Med Chem. 2020;63(3):905–927. DOI:10.1021/acs.jmed
chem.9b00835
30. Sun W, Chen LN, Zhou Q, et al. A unique hormonal recognition
feature of the human glucagon-like peptide-2 receptor. Cell Res.
2020;30(12):1098–1108. DOI:10.1038/s41422-020-00442-0
31. Norona J, Apostolova P, Schmidt D, et al. Glucagon-like peptide 2
for intestinal stem cell and Paneth cell repair during graft-versus-
host disease in mice and humans. Blood. 2020;136(12):1442–1455.
DOI:10.1182/blood.2020005957
32. Gattex FDA approval history. Available from: http://www.drugs.
com/history/gattex.html.04/03/2023
33. Gigola F, Cianci MC, Cirocchi R, et al. Use of Teduglutide in Children
with Intestinal Failure: a Systematic Review. Front Nutr. 2022 Jun
14;9:866518. DOI:10.3389/fnut.2022.866518
34. Coletta R, Morabito A, Iyer K. Nontransplant surgery for intestinal
failure. Gastroenterol Clin North Am. 2019;48(4):565–574.
35. Mundi MS, Mohamed Elfadil O, Hurt RT, et al. Management of
long-term home parenteral nutrition: historical perspective, com-
mon complications, and patient education and training. JPEN
J Parenter Enteral Nutr. 2023;47(1):S24–S34. DOI:10.1002/jpen.2424
36. van den Bosch CH, Jeremiasse B, van der Bruggen JT, et al. The
efficacy of taurolidine containing lock solutions for the prevention
of central-venous-catheter-related bloodstream infections:
a systematic review and meta-analysis. J Hosp Infect.
2021;123:143–155.
37. Mundi MS, Martindale RG, Hurt RT. Emergence of mixed-oil fat
emulsions for use in parenteral nutrition. JPEN J Parenter Enter
Nutr. 2017;41:3S–13S.
38. Chen YC, Chou CM, Huang SY, et al. Home parenteral nutrition for
children: what are the factors indicating dependence and
mortality? Nutrients. 2023;15(3):706. DOI:10.3390/nu15030706
39. Boroni G, Parolini F, Stern MV, et al. Autologous intestinal recon-
struction surgery in short bowel syndrome: which, When, and Why.
Front Nutr. 2022;9:861093.
40. Grainger JT, Maeda Y, Donnelly SC, et al. Assessment and manage-
ment of patients with intestinal failure: a multidisciplinary
approach. Clin Exp Gastroenterol. 2018;11:233–241.
41. Carey S, Kalachov M, Jones L, et al. Intestinal Failure Alliance of
Australia. Development of consensus on models of care in adults
with intestinal failure using a modified Delphi approach.
J Gastroenterol Hepatol. 33(9). Epub ahead of print. 1598–1602.
2018 Feb 27. 10.1111/jgh.14132.
42. Neelis E, de Koning B, van Winckel M, et al. Wide variation in
organization and clinical practice of pediatric intestinal failure
teams: an international survey. Clin Nutr. 2018;37:2271–2279.
43. Javid PJ, Wendel D, Horslen SP. Organization and outcomes of
multidisciplinary intestinal failure teams. Semin Pediatr Surg.
2018;27(4):218–222.
44. Merras-Salmio L, Mutanen A, Ylinen E, et al. Pediatric intestinal
failure: the key outcomes for the first 100 patients treated in
a National Tertiary Referral Center during 1984–2017. JPEN
J Parenter Enteral Nutr. 2018;42:1304–1313.
45. Abu-Elmagd KM, Armanyous SR, Fujiki M, et al. Management of five
hundred patients with gut failure at a single center: surgical inno-
vation versus transplantation with a novel predictive model. Ann
Surg. 2019;270(4):656–674. DOI:10.1097/SLA.0000000000003523
•• Historical paper on intestinal and multivisceral transplantation
by Pittsburgh group leaded by Abu-Elmagd KM
46. Fernandes MA, Usatin D, Allen IE, et al. Improved enteral tolerance
following step procedure: systematic literature review and
meta-analysis. Pediatr Surg Int. 2016;32(10):921–926. DOI:10.1007/
s00383-016-3927-9
47. International Step Data Registry (Boston Children’s Hospital at.
https://apps.childrenshospital.org/externalClinical/STEP/07/03/
2023
48. Lauro A, Santoro A, Cirocchi R, et al. Serial transverse enteroplasty
(STEP) in case of short bowel syndrome: did we achieve our goal?
A systematic review. Updates Surg. 2022;74(4):1209–1223. DOI:10.
1007/s13304-022-01316-3
49. Wester T, Lilja HE, Stenström P, et al. Absent ileocecal valve predicts
the need for repeated step in children. Surgery. 2017;161
(3):818–822. DOI:10.1016/j.surg.2016.09.011
50. Barrett M, Demehri FR, Ives GC, et al. Taking a STEP back: assessing
the outcomes of multiple STEP procedures. J Pediatr Surg. 2017;52
(1):69–73. DOI:10.1016/j.jpedsurg.2016.10.024
51. Coletta R, Aldeiri B, Morabito A. Institutional experience with spiral
intestinal lengthening and tailoring. Eur J Pediatr Surg. 2019;29
(5):412–416. DOI:10.1055/s-0038-1660850
First paper on SILT technique
52. Ripoli MC, Lauro A. Letter to the Editor on: “Gastrointestinal tract
reconstruction in adults with ultra-short-bowel syndrome: surgical
and nutritional outcomes”. Surgery. 2021;170(2):646.
53. van Praagh JB, Hofker HS, Haveman JW, et al. Comparing bowel
lengthening procedures: which, when, and why? Curr Opin Organ
Transplant. 2022;27(2):112–118.
54. Pironi L, Sasdelli AS. New insights into the indications for intestinal
transplantation. Curr Opin Organ Transplant. 2021;26:186–191.
•• New indications for intestinal transplantation by a worldwide
well-known leader on this field
55. Kaufman SS, Avitzur Y, Beath SV, et al. New Insights into the
Indications for Intestinal Transplantation: consensus in the Year
2019. Transplantation. 2020;104(5):937–946. DOI:10.1097/TP.
0000000000003065
56. Centers for Medicare & Medicaid Services - Intestinal and Multi-
Visceral Transplantation. https://www.cms.gov/medicare-coverage-
database/view. jul 3 2023
57. Kahn AB, Tulla KA, Tzvetanov IG. Indications of Intestinal
Transplantation. Gastroenterol Clin North Am. 2019;48(4):575–583.
58. Roberts AJ, Wales PW, Beath SV, et al. An international multicentre
validation study of the Toronto listing criteria for pediatric intest-
inal transplantion. Am J Transplant. 2022 Nov;22:2608–2615.
59. Smith JM, Weaver T, Skeans MA, et al. OPTN/SRTR 2018 Annual
Data Report: intestine. Am J Transplant. 2020;20:300–339.
60. Pironi L, Arends J, Bozzetti F, et al.; Home Artificial Nutrition &
Chronic Intestinal Failure Special Interest Group of ESPEN. ESPEN
guidelines on chronic intestinal failure in adults. Clin Nutr. 2016;35
(2):247–307. DOI:10.1016/j.clnu.2016.01.020
•• ESPEN guidelines on chronic intestinal failure in adults
61. Horslen SP, Smith JM, Weaver T, et al. OPTN/SRTR 2020 Annual
Data Report: intestine. Am J Transplant. 2022;22:310–349.
62. Lauro A, Marino IR, Iyer KR. Pre-emptive intestinal transplant: the
surgeon’s point of view. Dig Dis Sci. 2017;62(11):2966–2976. DOI:10.
1007/s10620-017-4752-2
Interesting paper on pre-emptive intestinal transplant written
by a worldwide expert
63. Talayero P, Ramos Boluda E, Gómez Massa E, et al. Donor-specific
antibodies in pediatric intestinal and multivisceral transplantation:
the role of liver and human leukocyte antigen mismatching. Liver
Transpl. 2018;24(12):1726–1735. DOI:10.1002/lt.25323
64. Fernández J, Bert F, Nicolas-Chanoine MH. The challenges of
multi-drug-resistance in hepatology. J Hepatol. 2016;65
(5):1043–1054.
65. Smith JM, Weaver T, Skeans MA, et al. OPTN/SRTR 2017 Annual
Data Report: intestine. Am J Transplant. 2019;19:284–322.
66. Bharadwaj S, Tandon P, Gohel TD, et al. Current status of intestinal
and multivisceral transplantation. Gastroenterol Rep. 2017;5:20–28.
67. Kesseli S, Sudan D. Small Bowel Transplantation. Surg Clin North
Am. 2019;99(1):103–116.
68. Matsumoto CS, Subramanian S, Fishbein TM. Adult intestinal
transplantation. Gastroenterol Clin North Am. 2018;47(2):341–354.
69. Farinelli PA, Rubio JS, Padín JM, et al. Use of nonvascularized
abdominal rectus fascia after liver, small bowel, and multiorgan
transplantation: long-term follow-up of a single-center series.
Transplant Proc. 2017;49(8):1810–1814. DOI:10.1016/j.transpro
ceed.2017.05.012
688 N. ZORZETTI ET AL.
70. Jo SC, McCallum Z, Shalley H, et al. Outcomes of children with
chronic intestinal failure: experience over 2 decades at a tertiary
pediatric hospital. J Pediatr Gastroenterol Nutr. 2019;69:79–87.
71. Norsa L, Artru S, Lambe C, et al. Long term outcomes of intestinal
rehabilitation in children with neonatal very short bowel syndrome:
parenteral nutrition or intestinal transplantation. Clin Nutr. 2019;38
(2):926–933. DOI:10.1016/j.clnu.2018.02.004
72. Sudan DL, Iyer KR, Deroover A, et al. A new technique for com-
bined liver/small intestinal transplantation. Transplantation.
2001;72(11):1846–1848. DOI:10.1097/00007890-200112150-00025
73. Fishbein T, Florman S, Gondolesi G, et al. Noncomposite simulta-
neous liver and intestinal transplantation. Transplantation. 2003;75
(4):564–565. DOI:10.1097/01.TP.0000045711.68765.84
74. Hukkinen M, Kivisaari R, Lohi J, et al. Transient elastography and
aspartate aminotransferase to platelet ratio predict liver injury in
paediatric intestinal failure. Liver Int. 2016;36(3):361–369. DOI:10.
1111/liv.12887
75. Beath SV, Davies P, Mukherjee A, et al. A comparison of two
validated scores for estimating risk of mortality of children with
intestinal failure associated liver disease and those with liver dis-
ease awaiting transplantation. Clin Res Hepatol Gastroenterol.
2014;38(1):32–39. DOI:10.1016/j.clinre.2013.06.001
76. Van Gossum A, Pironi L, Messing B, et al. Transient elastography (fibros-
can) is not correlated with liver fibrosis but with cholestasis in patients
with long-term home parenteral nutrition. JPEN J Parenter Enteral Nutr.
2015;39(6):719–724. DOI:10.1177/0148607114538057
77. Kato T, Nishida S, Levi D, et al. Multivisceral transplantation without
the liver. Transplant Proc. 2002;34(3):910. DOI:10.1016/S0041-
1345(02)02664-7
78. Vianna R, Beduschi T. Multivisceral transplantation for diffuse
splanchnic venous thrombosis. Curr Opin Organ Transplant.
2016;21(2):201–208.
79. Nikeghbalian S, Aliakbarian M, Shamsaeefar A, et al. Multivisceral
transplantation for the treatment of intra-abdominal tumors.
Transplant Proc. 2013;45(10):3528–3530. DOI:10.1016/j.transpro
ceed.2013.09.002
80. Matsumoto C, Kaufman S, Fishbein T. Inclusion of the colon in
intestinal transplantation. Curr Opin Organ Transplant. 2011;16
(3):312–315.
81. Moon JI, Zhang H, Waldron L, et al. “Stoma or no stoma”: first
report of intestinal transplantation without stoma. Am J Transplant.
2020;20(12):3550–3557. DOI:10.1111/ajt.16065
82. Hakim B, Myers DT, Williams TR, et al. Intestinal transplants: review
of normal imaging appearance and complications. Br J Radiol
Suppl. 2018;91(1090):20180173. DOI:10.1259/bjr.20180173
83. Grant D, Abu-Elmagd K, Mazariegos G, et al. Intestinal Transplant
Association. Intestinal transplant registry report: global activity and
trends. Am J Transplant. 2015;15(1):210–219. DOI:10.1111/ajt.12979
84. Gondolesi GE. Induction in intestinal transplantation.
Transplantation. 2020;104(10):1999–2000.
85. Elsabbagh AM, Hawksworth J, Khan KM, et al. Long-term survival in
visceral transplant recipients in the new era: a single-center experience.
Am J Transplant. 2019;19(7):2077–2091. DOI:10.1111/ajt.15269
86. Ceulemans LJ, Braza F, Monbaliu D, et al. The Leuven immunomo-
dulatory protocol promotes T-regulatory cells and substantially
prolongs survival after first intestinal transplantation. Am
J Transplant. 2016;16(10):2973–2985. DOI:10.1111/ajt.13815
87. Issa F, Strober S, Leventhal JR, et al. The fourth international work-
shop on clinical transplant tolerance. Am J Transplant. 2021;21
(1):21–31. DOI:10.1111/ajt.16139
88. Fu J, Zuber J, Shonts B, et al. Lymphohematopoietic graft-versus-
host responses promote mixed chimerism in patients receiving
intestinal transplantation. J Clin Invest. 2021;131(8):141698.
DOI:10.1172/JCI141698
89. Merola J, Shamim A, Weiner J. Update on immunosuppressive
strategies in intestinal transplantation. Curr Opin Organ
Transplant. 2022;27(2):119–125.
90. Santeusanio AD, Gu A, Weinberg AD, et al. Tacrolimus time-in-
therapeutic range is associated with freedom from acute rejection
and graft failure following intestinal transplantation. Clin
Transplant. 2021;35(6):14291. DOI:10.1111/ctr.14291
91. Andres AM, Talayero P, Alcolea Sanchez A, et al. Delayed introduc-
tion of sirolimus in paediatric intestinal transplant recipients: indi-
cations and long term benefits. Transplant Int. 2021;34:1895–1907.
92. Nishida S, Levi DM, Moon JI, et al. Intestinal transplantation with
alemtuzumab (Campath-1H) induction for adult patients.
Transplant Proc. 2006;38(6):1747–1749. DOI:10.1016/j.transpro
ceed.2006.05.057
93. Reyes J, Mazariegos GV, Abu-Elmagd K, et al. Intestinal transplanta-
tion under tacrolimus monotherapy after perioperative lymphoid
depletion with rabbit anti-thymocyte globulin (thymoglobulin). Am
J Transplant. 2005;5:1430–1436.
94. Vianna RM, Mangus RS, Fridell JA, et al. Induction immunosuppres-
sion with thymoglobulin and rituximab in intestinal and multivisc-
eral transplantation. Transplantation. 2008;85(9):1290–1293.
DOI:10.1097/TP.0b013e31816dd450
First paper on induction in intestinal and multivisceral trans-
plantation by thymoglobulin and rituximab
95. Beiras-Fernandez A, Chappell D, Hammer C, et al. Influence of
polyclonal anti-thymocyte globulins upon ischemia–reperfusion
injury in a non-human primate model. Transpl Immunol. 2006;15
(4):273–279. DOI:10.1016/j.trim.2006.02.003
96. Bogetti D, Jarzembowski TM, Sankary HN, et al. Hepatic ischemia/
reperfusion injury can be modulated with thymoglobulin induction
therapy. Transplant Proc. 2005;37(1):404–406. DOI:10.1016/j.trans
proceed.2004.12.064
97. Vianna R, Farag A, Gaynor JJ, et al. Association of More Intensive
Induction with Less Acute Rejection Following Intestinal
Transplantation: results of 445 Consecutive Cases from a Single
Center. Transplantation. 2020;104(10):2166–2178. DOI:10.1097/TP.
0000000000003074
98. Abu-Elmagd KM, Wu G, Costa G, et al. Preformed and de novo
donor specific antibodies in visceral transplantation: long-term
outcome with special reference to the liver. Am J Transplant.
2012;12(11):3047–3060. DOI:10.1111/j.1600-6143.2012.04237.x
•• Outstanding paper on the role of DSA in intestinal and multi-
visceral transplantation by a leading expert in the field
99. Cheng EY, Everly MJ, Kaneku H, et al. Prevalence and clinical impact
of donor specific alloantibody among intestinal transplant
recipients. Transplantation. 2017;101:873–882.
100. Gerlach UA, Lachmann N, Ranucci G, et al. Non-HLA antibodies may
accelerate immune responses after intestinal and multivisceral
transplantation. Transplantation. 2017;101(1):141–149. DOI:10.
1097/TP.0000000000001439
Interesting paper on the role of nonHLA antibodies in intest-
inal and multivisceral transplantation
101. Santeusanio AD, Moon J, Nair V, et al. Is There a Role for
Desensitization in Intestinal Transplantation? Prog Transplant.
2019;29(3):275–278. DOI:10.1177/1526924819855088
102. Garcia-Roca R, Tzvetanov IG, Jeon H, et al. Successful living donor
intestinal transplantation in cross-match positive recipients: initial
experience. World J Gastrointest Surg. 2016;8:101–105.
103. Horslen SP, Smith JM, Ahn Y, et al. OPTN/SRTR 2019 annual data
report: intestine. Am J Transplant. 2021;21:316–355.
104. Huard G, Schiano TD, Moon J, et al. Severe acute cellular rejection
after intestinal transplantation is associated with poor patient and
graft survival. Clin Transplant. 2017;31(5):12956. DOI:10.1111/ctr.
12956
105. Devine K, Ranganathan S, Mazariegos G, et al. Induction regimens
and posttransplantation lymphoproliferative disorder after pedia-
tric intestinal transplantation: single-center experience. Pediatr
Transplant. 2020;24:13723.
106. Wu GS, Zhao QC, Li ZS, et al. Successful rescue of late-onset anti-
body mediated rejection 12 years after living-donor intestinal
transplantation: a case report. Transplant Proc. 2017;49:232–236.
107. Trentadue G, Kats-Ugurlu G, Blokzijl T, et al. Safe and successful
treatment of acute cellular rejection of an intestine and abdominal
wall transplant with vedolizumab. Transplant Direct. 2020;6:527.
EXPERT REVIEW OF GASTROENTEROLOGY & HEPATOLOGY 689
108. Kroemer A, Belyayev L, Khan K, et al. Rejection of intestinal allo-
transplants is driven by memory T helper type 17 immunity and
responds to infliximab. Am J Transplant. 2021;21(3):1238–1254.
DOI:10.1111/ajt.16283
109. Sano T, Kageyama T, Fang V, et al. Redundant cytokine require-
ment for intestinal microbiota-induced Th17 cell differentiation in
draining lymph nodes. Cell Rep. 2021;36(8):109608. DOI:10.1016/j.
celrep.2021.109608
110. Fujiwara S, Wada M, Kudo H, et al. Effectiveness of bortezomib in
a patient with acute rejection associated with an elevation of
donor-specific HLA antibodies after small-bowel transplantation:
case report. Transplant Proc. 2016;48:525–527.
111. Fan J, Tryphonopoulos P, Tekin A, et al. Eculizumab salvage therapy
for antibody-mediated rejection in a desensitization-resistant
intestinal re-transplant patient. Am J Transplant. 2015;15
(7):1995–2000. DOI:10.1111/ajt.13183
112. Cristofori F, Dargenio VN, Dargenio C, et al. Anti-inflammatory and
immunomodulatory effects of probiotics in gut inflammation:
a door to the body. Front Immunol. 2021;12:578386.
113. Burrello C, Garavaglia F, Cribiù FM, et al. Therapeutic faecal micro-
biota transplantation controls intestinal inflammation through IL10
secretion by immune cells. Nat Commun. 2018;9(1):5184. DOI:10.
1038/s41467-018-07359-8
114. Issa NC, Fishman JA. Infectious complications of antilymphocyte
therapies in solid organ transplantation. Clin Infect Dis. 2009;48
(6):772–786.
115. Kubal CA, Pennington K, Fridell J, et al. Challenges with Intestine
and Multivisceral Re-Transplantation: importance of Timing of
Re-Transplantation and Optimal Immunosuppression. Ann
Transplant. 2018;23:98–104.
116. Kroemer A, Khan K, Kaufman SS, et al. Operational tolerance in
intestinal transplantation. Am J Transplant. 2021;21(2):876–882.
DOI:10.1111/ajt.16224
117. Clouse JW, Kubal CA, Fridell JA, et al. One-year Incidence of
Infection in Pediatric Intestine Transplantation. Pediatr Infect Dis
J. 2019;38(3):219–223. DOI:10.1097/INF.0000000000002104
118. Fishbein TM. Intestinal transplantation. N Engl J Med. 2009;361
(10):998–1008.
119. Oertel SH, Verschuuren E, Reinke P, et al. Effect of anti-CD 20
antibody rituximab in patients with post-transplant lymphoproli-
ferative disorder (PTLD). Am J Transplant. 2005;5:2901–2906.
120. Kaenkumchorn TK, Wendel D, Pacheco MC, et al. Late graft loss
after intestinal transplantation. Curr Opin Organ Transplant.
2021;26(2):220–228. DOI:10.1097/MOT.0000000000000851
121. Ganoza A, Mazariegos GV, Khanna A. Current status of graft-versus-
host disease after intestinal transplantation. Curr Opin Organ
Transplant. 2019;24(2):199–206.
122. Kaufman SS, Hussan E, Kroemer A, et al. Graft versus Host Disease
After Intestinal Transplantation: a Single-center Experience.
Transplant Direct. 2021;7:731.
123. Vianna R, Farag A, Gaynor JJ, et al. Association of Alemtuzumab
Induction with a Significantly Lower Incidence of GVHD Following
Intestinal Transplantation: results of 445 Consecutive Cases from
a Single Center. Transplantation. 2020;104(10):2179–2188. DOI:10.
1097/TP.0000000000003111
Outstanding series on Alemtuzumab role in reducing GVHD
after intestinal transplantation
124. Venick RS. Long-term results of intestinal transplantation in chil-
dren: survival after 10 years, intestinal function, quality of life. Curr
Opin Organ Transplant. 2018;23(2):219–223.
125. Gondolesi GE, Fernandez A, Burghardt KM, et al. Meeting report of
the XIV international small bowel transplant symposium: summary
of presentations, workshops, and debates from a comprehensive
meeting on intestinal failure, rehabilitation, and transplantation,
Buenos Aires, Argentina, June 10-13, 2015. JPEN J Parenter
Enteral Nutr. 2018;42:477–489.
126. Garcia J, Selvaggi G, Tekin A, et al. Intestinal transplantation. Curr
Opin Organ Transplant. 2021;26(2):229–233. DOI:10.1097/MOT.
0000000000000865
127. Oltean M. Intestinal transplantation: an overview of the recent experi-
mental studies. Curr Opin Organ Transplant. 2021;26(2):240–244.
128. Zou Z, Wang Z. Liraglutide attenuates intestinal ischemia/reperfu-
sion injury via NF-κB and PI3K/Akt pathways in mice. Life Sci.
2022;309:121045.
129. Lauro A, Marino IR, Matsumoto CS. Advances in allograft monitor-
ing after intestinal transplantation. Curr Opin Organ Transplant.
2016;21(2):165–170.
130. Carroll RE. Endoscopic follow-up of intestinal transplant recipient.
Gastroentrol Clin North Am. 2018;47:381–391.
131. Varkey J, Stotzer PO, Simren M, et al. The endoscopic surveillance
of the transplanted small intestine: a single center experience and
a proposal for a grading score. Scand J Gastroenterol. 2018;53
(2):134–139. DOI:10.1080/00365521.2017.1411523
132. Gerlach UA, Klöpfel M, Atanasov G, et al. Intragraft and Systemic
Immune Parameters Discriminating Between Rejection and
Long-Term Graft Function in a Preclinical Model of Intestinal
Transplantation. Transplantation. 2017;101(5):1036–1045. DOI:10.
1097/TP.0000000000001469
133. Osman M, Parekh N, Fujiki M, et al. Disease Recurrence after Gut
Transplantation. Curr Opin Organ Transplant. 2021;26(2):207–219.
DOI:10.1097/MOT.0000000000000856
134. Hind J. Long-term outcomes of intestinal transplantation. Curr Opin
Organ Transplant. 2021;26(2):192–199.
135. Oltean M, Hedenström P, Varkey J, et al. Endoscopic ultrasound in
the monitoring of the intestinal allograft. BMJ Open Gastroenterol.
2022;9(1):e000792. DOI:10.1136/bmjgast-2021-000792
136. Pironi L, Guidetti M, Lauro A, et al. Plasma citrulline after small
bowel transplantation: effect of time from transplantation, acute
cellular rejection, and renal failure. Clin Transplant. 2015;29
(11):1039–1046. DOI:10.1111/ctr.12630
137. Smullin CP, Venick RS, Marcus EA, et al. REG3α is a novel biomarker that
potentially correlates with acute allograft rejection after intestinal trans-
plantation. Clin Transplant. 2021;35(7):e14378. DOI:10.1111/ctr.14378
138. Guerra MR, Rossetti M, Zhang Z, et al. Characterization of T cell
immunophenotypes in intestinal transplantation: a pilot study.
Transpl Immunol. 2018;51:50–57.
139. Tran LC, Lazonby G, Morello R, et al. How good is quality-of-life for
children receiving home parenteral nutrition? - a pilot study. Clin
Nutr ESPEN. 2019;29:119–124.
140. Baxter JP, Fayers PM, Bozzetti F, et al. Home Artificial Nutrition and
Chronic Intestinal Failure Special Interest Group of the European
Society for Clinical Nutrition and Metabolism (ESPEN). An interna-
tional study of the quality of life of adult patients treated with
home parenteral nutrition. Clin Nutr. 2019;38(4):1788–1796. DOI:10.
1016/j.clnu.2018.07.024
141. Roskott AM, Groen H, Rings EH, et al. Cost-effectiveness of intest-
inal transplantation for adult patients with intestinal failure:
a simulation study. Am J Clin Nutr. 2015;101(1):79–86. DOI:10.
3945/ajcn.114.083303
142. Raghu VK, Rudolph JA, Smith KJ. Cost-effectiveness of teduglutide
in pediatric patients with short bowel syndrome: Markov modeling
using traditional cost-effectiveness criteria. Am J Clin Nutr.
2021;113(1):172–178.
143. Hussain S, Mehta A, Pettle T, et al. Multicenter Evaluation of Current
Knowledge of Intestinal Transplantation and Rehabilitation Among
Trainee Physicians. Transplant Proc. 2015;47(6):1988–1992. DOI:10.
1016/j.transproceed.2015.04.099
144. Iyer K, Moon J. Adult intestinal transplantation in the United States.
Curr Opin Organ Transplant. 2020;25(2):196–200.
145. Kaplan J, Han L, Halgrimson W, et al. The impact of MELD/PELD
revisions on the mortality of liver-intestine transplantation
candidates. Am J Transplant. 2011;11:1896–1904.
690 N. ZORZETTI ET AL.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Parenteral nutrition (PN) in children with short bowel syndrome is crucial and lifesaving. Taking care of such patients requires interprofessional practice and multiple team resource management. Home PN (HPN) usage allows patients and families to live regular lives outside hospitals. We share our experiences for the last two decades and identify the risk factors for complications and mortality. A retrospective study of HPN patients was conducted between January 2000 and February 2022. Medical records of age, body weight, diagnosis, length of residual intestines, HPN period, central line attempts, complications, weaning, and survival were collected and analyzed. The patients were classified as HPN free, HPN dependent, and mortality groups. A total of 25 patients received HPN at our outpatient clinic, and one was excluded for the adult age of disease onset. There were 13 patients (54.1%) who were successfully weaned from HPN until the record-enroled date. The overall mortality rate was 20.8% (five patients). All mortality cases had prolonged cholestasis, Child Class B or C, and a positive Pediatric End-Stage Liver Disease (PELD) score. For HPN dependence, extended resection and multiple central line placement were two significant independent factors. Cholestasis, Child Class B or C, and positive PELD score were the most important risk factors for mortality. The central line-related complication rate was not different in all patient groups. The overall central line infection rate was 1.58 per 1000 catheter days. Caution should be addressed to prevent cholestasis and intestinal failure-associated liver disease during the HPN period, to prevent mortality. By understanding the risks of HPN dependence and mortality, preventive procedures could be addressed earlier.
Article
Full-text available
While the history of nutrition support dates to the ancient world, modern home parenteral and enteral nutrition (HPEN) has been available since the 1960s. Home enteral nutrition is primarily for patients in whom there is a reduction in oral intake below the amount needed to maintain nutrition or hydration (i.e., oral failure), whereas home parenteral nutrition is used for patients when oral-enteral nutrition is temporarily or permanently impossible or absorption insufficient to maintain nutrition or hydration (i.e., intestinal failure). The development of home delivery of these therapies has revolutionized the field of clinical nutrition. The use of HPEN appears to be increasing on a global scale, and because of this, it is important for healthcare providers to understand all that HPEN entails to provide safe, efficacious, and cost-effective support to the HPEN patient. In this article, we provide a comprehensive review of the indications, patient requirements, monitoring, complications, and overall process of managing these therapies at home. Whereas some of the information in this article may be applicable to the pediatric patient, the focus is on the adult population.
Article
Full-text available
Surveys on Serial Transverse Enteroplasty (STEP) published in international literature (1 January 2003– 31 May 2021) were searched. Articles were included from 17 countries: 1/23 comparative and 22/23 cohort studies. STEP was performed on 308 patients: pediatrics, adults, and mixed ages. Pediatric group included 16 studies and the adult 6. Pre-STEP residual small bowell (SB) length for pediatrics and adults ranged from 18 to 26 cm and from 30 to 70 cm, respectively. Post-STEP increased SB length for pediatrics and adults ranged between 42 and 100% and 50% and 176%, respectively. For pediatrics, enteral autonomy was reached in 32.22% of cases, parenteral nutrition (PN) dependence was 36.11%, a repeated STEP procedure (Re-STEP) was needed in 17.22%, and a bowel transplant was performed in 6.11%. In adults, enteral autonomy was achieved in 52.38%, while PN dependence was 37.1%, and no Re-STEP or transplantation were required. For the mixed group, post-STEP bowel length increased from 2 to 50 cm, enteral autonomy was obtained in 43%, PN dependence was 57%, without reported Re-STEP or transplantation. Mortality rates were between 5.55% (pediatric) and 7.14% (adults). Preoperative length with preservation of ileocecal valve represented the main predictive factors to achieve enteral autonomy.
Article
Full-text available
Background and Objectives Short-bowel syndrome (SBS) results from the loss of a significant portion of the small intestine leading to a state of malabsorption. After an intestinal loss, there is a process of adaptation involving the Glucagon-Like Peptide-2 (GLP-2), an enteroendocrine peptide also involved in nutrient absorption. Teduglutide is a recombinant analog of GLP-2 approved in 2016 to treat selected SBS pediatric patients who are dependent on parenteral support. The present systematic review aims to evaluate the efficacy of Teduglutide in pediatric patients with SBS in reducing the need for parenteral nutrition (PN). Materials and Methods We performed a literature search on MEDLINE and Embase to include articles up to November 2021. We included articles that involved using Teduglutide in the SBS pediatric population to define its efficacy in reducing the need for PN. The key words used were GLP-2, teduglutide, child. Results Fourteen studies completely fulfilled the inclusion criteria. Two hundred 23 patients were treated with Teduglutide, and the median duration of treatment was 45 weeks (IQR: 36–52.5 weeks). One-hundred and fifty-two patients were treated with 0.05 mg/Kg/d of subcutaneous Teduglutide, 38 received 0.025 mg/Kg/d and 8 received either 0.125 mg/Kg/d or 0.20 mg/Kg/d. A total of 36 patients achieved enteral autonomy (EA) after a median of 24 weeks of treatment (IQR: 24–48 weeks) and 149 patients showed a reduction in PN needs in terms of volume, calories, or hours per day. Eleven studies reported complications: gastrointestinal were the most common, with 89 cases reported in treated patients and 11 in non-treated patients. Conclusion Teduglutide appears safe and effective in reducing PN requirements and improving EA in the pediatric population. However, more studies are needed to understand its efficacy in the long term and after discontinuation and possible complications. Systematic Review Registration [https://www.crd.york.ac.uk/prospero/], identifier [CRD42022301593].
Article
Full-text available
Background and Aims Chronic intestinal failure (CIF) therapy changed significantly in recent decades, and both survival and complication rates improved over time. International guidelines claim that early referral of long-term home parenteral nutrition (HPN) patients to an expert center with specific standards of care may positively affect long-term outcomes. Herein, we retrospectively analyse the long-term outcomes of a cohort of pediatric patients with CIF followed-up since our Pediatric Intestinal Failure Unit foundation, in 1989. Methods Data of the 120 children followed up at Pediatric Intestinal Failure Unit during the last 28 years were retrospectively collected. Patients' and HPN characteristics, as well as dependence, survival, and complication rates, were described. Results Incidence and prevalence of CIF increased during the study period particularly due to the increase of HPN for non-digestive disease (NDD) CIF (47.5% of the study sample). Catheter-related bloodstream infection (CRBSI) rate decreased over the study period: 0.33 episodes/1,000 catheters days before 2011 and 0.19 episodes/1,000 catheters days afterwards. Only 1 patient out of 12 died because of HPN complications. The survival rate of patients with PDD was 98.4% at 1 year from the beginning of HPN, 96.5% at 2 years, and 93.8% from the fifth year onwards. Concerning the dependence rate, 70.6% of patients were still on HPN 1 year after the start of HPN, 63.7% at 2 years, 52.4% at 5 years, and 40.8% from the 9th year onwards, with no significant difference according to the underlying intestinal pathology. The survival rate of NDD patients was 91.2% at 1 year from the beginning of HPN, 87.4% at 2 years, and 81.9% from the third year onwards. For what concerns the enteral autonomy, it was regained by 56.7% 1 year after the start of HPN, 74.5% at 2 years, and 95.0% in the 5th year. Conclusions Our data confirmed the importance of appropriate standards of care and suggest that applying a specific set of standards and protocols may further improve patients' outcomes and survival. Indeed, both primary and non-digestive diseases HPN showed good outcomes.
Article
Intestinal failure (IF) is a rare disease that requires ongoing intravenous supplementation to sustain growth and health. Advancements in parenteral nutrition (PN) and central venous access in the 1960s and 1970s transformed a life‐limiting disease to a chronic one in which patients are able to administer hydration, electrolytes, micronutrients, and macronutrients in the comfort of their home. However, despite ongoing advancements in the field of home PN (HPN), complications—whether related to central venous catheters or PN itself—remain common and can be associated with significant morbidity and mortality. Central venous access can be associated with thrombosis, central line–associated bloodstream infection, or damage and can result in loss of access over time. PN can be associated with IF‐associated liver disease or hyperglycemia. The key to preserving central venous access and quality of life and maintaining health for patients with chronic IF (CIF) is education focused on prevention and prompt management of CIF complications as they arise. This education typically takes place at the time of initiation of HPN, either in the hospital setting or in the patient's home. The present manuscript describes the historical progression of HPN, prevalence and characteristics of CIF, and an in‐depth discussion of the most common catheter‐related and PN‐related complications and their management, along with a discussion of our education and training process.
Article
Aims Previous studies have reported that glucagon-like peptide-1 (GLP-1) may play a critical role in the development of intestinal ischemia-reperfusion (I/R) injury. The present study aimed to investigate whether liraglutide (GLP-1 analog) protects against intestinal I/R injury and reveals the possible underlying mechanism. Main methods Temporary superior mesenteric artery occlusion was performed to establish an intestinal I/R injury mouse model. Different doses of liraglutide were administered in vivo. Then, the survival rate of mice exposed to different ischemia times, the histopathology, intestinal barrier index, cytokine production, intestinal tissue apoptosis, and the levels of several proteins were detected in each group. Key findings Pretreatment with liraglutide significantly alleviated the pathological changes induced by I/R and increased the overall survival of mice exposed to intestinal I/R injury. Moreover, liraglutide attenuated neutrophil infiltration of intestinal tissues, pro-inflammatory cytokine production (including IL-1β, IL-6, and TNF-α), and apoptosis of intestinal tissues caused by intestinal I/R injury. In addition, liraglutide inhibited the nuclear translocation of nuclear factor-κB (NF-κB) and up-regulated the phosphorylation levels of phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt) in the I/R group. Significance Liraglutide may attenuate the inflammatory response and the apoptosis of intestinal tissues via the NF-κB and PI3K/Akt pathway, protecting against intestinal I/R.
Article
Background and aims: Patients with intestinal failure receiving home parenteral nutrition (HPN) are susceptible to central-line associated bloodstream infections (CLABSIs), with crucial roles for adequate (empiric) antimicrobial therapy and effective catheter management strategies. Our aim was to link recent epidemiologic CLABSI data with clinical outcomes and to identify risk factors for therapeutic failure to decide on the safest and most accurate CLABSI management in patients receiving HPN. Methods: A retrospective observational cohort study was conducted. All data on CLABSIs (period 2010-2020) in adult patients receiving HPN were retrieved. The efficacy of attempted catheter salvage and empiric antimicrobial treatment (β-lactam antibiotics) in our center, with a low prevalence of methicillin-resistant staphylococci, was investigated. Multivariate cox-regression analysis was performed to identify risk factors for recurrent CLABSI. Results: 389 CLABSIs occurred in 149 patients. The overall infection rate was 0.64 per 1000 central venous catheter (CVC) days. Most CLABSIs were caused by Coagulase-negative staphylococci (37%). Attempted CVC salvage was successful in 70% of the cases. Empiric antimicrobial therapy was found to be adequate in only 47% of cases, mainly because of insufficient Coagulase-negative staphylococci coverage. According to the Cox model, patients with a replaced CVC had a 50% lower risk of a new CLABSI than patients with a retained (salvaged) CVC during follow-up (HR 0.50; 95% CI 0.35-0.72, P < 0.001). Conclusions: CVC salvage can be achieved in most CLABSI cases but seems associated with a shorter CLABSI-free survival. Importantly, based on our findings, a glycopeptide containing antibiotic treatment regimen will increase the likelihood of adequate empiric coverage.
Article
Deciding which patients would benefit from intestinal transplantation (IT) remains an ethical/clinical dilemma. New criteria* were proposed in 2015: ≥2 intensive care unit (ICU) admissions, loss of ≥3 central venous catheter (CVC) sites, and persistently elevated conjugated bilirubin (CB ≥75μmol/L) despite 6 weeks of lipid modification strategies. We performed a retrospective, international, multicentre validation study of 443 children (61% male, median gestational age 34 weeks [IQR 29‐37]), diagnosed with IF between 2010 & 2015. Primary outcome measure was death or IT. Sensitivity, specificity, NPV, PPV, probability of death/transplant (OR, 95% confidence intervals) were calculated for each criterion. Median age at IF diagnosis was 0.1 years (IQR 0.03‐0.14) with median follow up of 3.8 years (IQR 2.3‐5.3). 40/443 (9%) patients died, 53/443 (12%) were transplanted; 11 died post‐ transplant. The validated criteria had a high predictive value of death/IT; ≥2 ICU admissions (p <0.0001, OR 10.2, 95% CI 4.0‐25.6), persistent CB ≥75μmol/L (p<0.0001, OR 8.2, 95% CI 4.8‐13.9) and loss of ≥3 CVC sites (p=0.0003, OR 5.7, 95% CI 2.2‐14.7). This large, multicentre, international study in a contemporary cohort confirms the validity of the Toronto criteria. These validated criteria should guide listing decisions in pediatric IT. *Burghadt KM et al. AJT 2015;15:1674‐81.
Article
Background Survival has frequently been studied and reported in patients with long term intestinal failure (IF). However, studies comparing the survival and mortality rates with the background population are rare. This study compares the survival in an adult IF, non-malignant, short bowel syndrome (SBS) cohort with a control group and with age- and sex-specific background mortality rates. Design Patients with SBS, defined by a small bowel length of 200 cm or less, due to non-malignant disease, were included and followed until death or censoring on 31 December 2017. Causes of deaths occurring during home parenteral support (HPS) were assessed by review of the charts. Each case was matched with ten controls from the background population according to year of HPS initiation, age, and sex, and their survival was compared. Furthermore, age- and sex-specific mortality rates of the background population were used to calculate the standardized mortality ratio (SMR) and excess mortality. Results After five years, patients who initiated HPS had a relative survival of 76%. The SMR was 5.0 and the excess mortality was 50 per 1,000 years. HPS-related deaths were assessed to account for 11% of deaths during HPS and occurred with an incidence of 10 per 1,000 years. The excess mortality was as low as 15 per 1,000 years in cases aged less than 40 years. Patients weaning off HPS had a mortality rate closer to that expected in the background population. Conclusions In adult patients with a non-malignant cause of SBS-IF, the excess mortality was 50 per 1,000 years. However, HPS related deaths were rarely registered with an incidence of 10 HPS related deaths per 1,000 HPS treatment years.