ArticlePDF Available

Brazilian guidelines for the management of brain-dead potential organ donors. The task force of the AMIB, ABTO, BRICNet, and the General Coordination of the National Transplant System

Authors:

Abstract and Figures

Objective To contribute to updating the recommendations for brain-dead potential organ donor management. Method A group of 27 experts, including intensivists, transplant coordinators, transplant surgeons, and epidemiologists, joined a task force formed by the General Coordination Office of the National Transplant System/Brazilian Ministry of Health (CGSNT-MS), the Brazilian Association of Intensive Care Medicine (AMIB), the Brazilian Association of Organ Transplantation (ABTO), and the Brazilian Research in Intensive Care Network (BRICNet). The questions were developed within the scope of the 2011 Brazilian Guidelines for Management of Adult Potential Multiple-Organ Deceased Donors. The topics were divided into mechanical ventilation, hemodynamic support, endocrine-metabolic management, infection, body temperature, blood transfusion, and use of checklists. The outcomes considered for decision-making were cardiac arrest, number of organs recovered or transplanted per donor, and graft function/survival. Rapid systematic reviews were conducted, and the quality of evidence of the recommendations was assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system. Two expert panels were held in November 2016 and February 2017 to classify the recommendations. A systematic review update was performed in June 2020, and the recommendations were reviewed through a Delphi process with the panelists between June and July 2020. Results A total of 19 recommendations were drawn from the expert panel. Of these, 7 were classified as strong (lung-protective ventilation strategy, vasopressors and combining arginine vasopressin to control blood pressure, antidiuretic hormones to control polyuria, serum potassium and magnesium control, and antibiotic use), 11 as weak (alveolar recruitment maneuvers, low-dose dopamine, low-dose corticosteroids, thyroid hormones, glycemic and serum sodium control, nutritional support, body temperature control or hypothermia, red blood cell transfusion, and goal-directed protocols), and 1 was considered a good clinical practice (volemic expansion). Conclusion Despite the agreement among panel members on most recommendations, the grade of recommendation was mostly weak. The observed lack of robust evidence on the topic highlights the importance of the present guideline to improve the management of brain-dead potential organ donors.
Content may be subject to copyright.
Westphaletal. Ann. Intensive Care (2020) 10:169
https://doi.org/10.1186/s13613-020-00787-0
RESEARCH
Brazilian guidelines forthemanagement
ofbrain-dead potential organ donors.
The task force oftheAMIB, ABTO, BRICNet,
andtheGeneral Coordination oftheNational
Transplant System
Glauco Adrieno Westphal1,2,3*, Caroline Cabral Robinson1, Alexandre Biasi Cavalcanti4,
Anderson Ricardo Roman Gonçalves5,6, Cátia Moreira Guterres1, Cassiano Teixeira7,8, Cinara Stein1,
Cristiano Augusto Franke7,9, Daiana Barbosa da Silva1, Daniela Ferreira Salomão Pontes10,
Diego Silva Leite Nunes10, Edson Abdala11, Felipe Dal‑Pizzol12,13, Fernando Augusto Bozza14,15,
Flávia Ribeiro Machado16, Joel de Andrade17, Luciane Nascimento Cruz1, Luciano Cesar Pontes de Azevedo18,
Miriam Cristine Vahl Machado3, Regis Goulart Rosa1, Roberto Ceratti Manfro7,19, Rosana Reis Nothen19,
Suzana Margareth Lobo20, Tatiana Helena Rech7, Thiago Lisboa7, Verônica Colpani1 and Maicon Falavigna1,21,22
Abstract
Objective: To contribute to updating the recommendations for brain‑dead potential organ donor management.
Method: A group of 27 experts, including intensivists, transplant coordinators, transplant surgeons, and epidemi‑
ologists, joined a task force formed by the General Coordination Office of the National Transplant System/Brazilian
Ministry of Health (CGSNT‑MS), the Brazilian Association of Intensive Care Medicine (AMIB), the Brazilian Association
of Organ Transplantation (ABTO), and the Brazilian Research in Intensive Care Network (BRICNet). The questions were
developed within the scope of the 2011 Brazilian Guidelines for Management of Adult Potential Multiple‑Organ
Deceased Donors. The topics were divided into mechanical ventilation, hemodynamic support, endocrine‑metabolic
management, infection, body temperature, blood transfusion, and use of checklists. The outcomes considered for
decision‑making were cardiac arrest, number of organs recovered or transplanted per donor, and graft function/sur‑
vival. Rapid systematic reviews were conducted, and the quality of evidence of the recommendations was assessed
using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system. Two expert
panels were held in November 2016 and February 2017 to classify the recommendations. A systematic review update
was performed in June 2020, and the recommendations were reviewed through a Delphi process with the panelists
between June and July 2020.
Results: A total of 19 recommendations were drawn from the expert panel. Of these, 7 were classified as strong
(lung‑protective ventilation strategy, vasopressors and combining arginine vasopressin to control blood pressure,
© The Author(s) 2020. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material
in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material
is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the
permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creat iveco
mmons .org/licen ses/by/4.0/.
Open Access
*Correspondence: glauco.ww@gmail.com.br
1 Hospital Moinhos de Vento (HMV), R. Ramiro Barcelos, 910, Porto Alegre,
RS 90035000, Brazil
Full list of author information is available at the end of the article
Page 2 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
Introduction
Organ donation for transplantation is a complex process
led by several health care professionals responsible for a
sequence of actions and procedures that begin with iden-
tifying a potential organ donor and end with organ pro-
curement surgery and distribution. e progress of this
process is essential to increase the deceased-donor pool,
and to decrease the growing disparity between the num-
ber of patients on transplant waiting lists and the avail-
ability of organs [1, 2].
e organ donation process includes the identification
of the potential donor, diagnosis of brain death, family
support and interview, evaluation of donor eligibility cri-
teria, clinical management of the potential organ donor,
and organ procurement and distribution [2, 3]. Given
the marked clinical instability that occurs in patients
who progress to brain death, the application of poten-
tial donor-management strategies aiming at hemody-
namic stabilization is crucial to avoid loss of organs due
to hypoperfusion or loss of donors due to cardiac arrest.
Also, the control of ventilatory support, body tempera-
ture, and endocrine-metabolic functions contributes to
improving the quality of organs and clinical outcomes in
transplant recipients [1, 2, 4, 5].
Despite the lack of evidence on some aspects of the
clinical management of potential organ donors, the
recommendations presented in this guideline intend
to promote a general approach to mitigate the dis-
parity between supply and demand of organs for
transplantation.
Objective
To provide recommendations to guide the clinical man-
agement of brain-dead potential organ donors aiming to
reduce the rate of cardiac arrest of the potential donor
and to improve organ viability for transplantation.
Method
e present document provides a partial update on the
2011 Brazilian Guidelines for Management of Adult
Potential Multiple-Organ Deceased Donors [68].
e working group consisted of physicians, nurses,
pharmacists, physical therapists, epidemiologists, meth-
odologists, and transplant system managers. e contri-
butions of each participant are shown in Additional file1,
and the respective conflict-of-interest disclosures are
shown in Additional file2.
e target audience of this guideline is health care pro-
fessionals, especially physicians and nursing staff work-
ing in adult ICUs and emergency departments, who are
involved in the care of adult individuals with known or
suspected brain death.
e clinical issues addressed by the guideline were
defined by coordinators of the working group and the
methodologists in a face-to-face meeting held in March
2016, after reviewing the recommendations of the 2011
Brazilian Guidelines for Management of Adult Potential
Multiple-Organ Deceased Donors [68]. e issues were
prioritized according to the perception of their impact on
medical management and variability in clinical practice
and divided into the following major topics: (1) ventila-
tory support; (2) hemodynamic support; (3) endocrine,
metabolic and nutritional management; (4) specific
aspects that include infection and sepsis, red blood cell
transfusion, and body temperature control; and (5) goal-
directed therapy. For each clinical issue, operational
questions were developed and framed using the PICO
(population-intervention-comparison-outcome) format.
e population of interest consists of potential organ
donors with known or suspected brain death [3], hereaf-
ter referred to as potential donors. e outcomes consid-
ered for decision-making were cardiac arrest, the number
of organs recovered or transplanted per donor, and graft
function or graft survival.
For each clinical issue, rapid systematic reviews [9, 10]
were conducted using the following search strategy: (1)
Review of the reference lists of Brazilian guidelines [68]
and the Society of Critical Care Medicine (SCCM) [11]
statement on the management of the potential organ
donor; (2) Review of related topics in the DynaMed and
UpToDate databases; and (3) PubMed search focusing
on systematic reviews and clinical trials published until
October 2016 and until January 2017. Quality of evidence
was assessed using the Grading of Recommendations
antidiuretic hormones to control polyuria, serum potassium and magnesium control, and antibiotic use), 11 as weak
(alveolar recruitment maneuvers, low‑dose dopamine, low‑dose corticosteroids, thyroid hormones, glycemic and
serum sodium control, nutritional support, body temperature control or hypothermia, red blood cell transfusion, and
goal‑directed protocols), and 1 was considered a good clinical practice (volemic expansion).
Conclusion: Despite the agreement among panel members on most recommendations, the grade of recommenda‑
tion was mostly weak. The observed lack of robust evidence on the topic highlights the importance of the present
guideline to improve the management of brain‑dead potential organ donors.
Keywords: Guidelines, Organ donation, Intensive care, Brain death, GRADE
Page 3 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
Assessment, Development, and Evaluation (GRADE) sys-
tem [12].
e recommendations were prepared and submitted to
two face-to-face expert panels held in November 2016,
and February 2017. For each recommendation, the direc-
tion of the course of action was discussed (whether to
perform or not to perform the proposed action), and the
strength of the recommendation was classified as strong
or weak according to the GRADE system [12]. After the
last panel meeting, a new systematic search covering the
period from October 2016 to May 2020 was carried out
to identify new evidence that could potentially modify
the recommendations. From June to July 2020, a Delphi
process was performed with the panelists to present the
results of the literature update and review the direction
and strength of the recommendations.
Results
A total of 19 recommendations were drawn from the
expert panel. Of these, 7 were classified as strong, 11 as
weak, and 1 was considered as good clinical practice.
Table1 shows a summary of the recommendations. Fig-
ure1 presents graphically the flow of the recommenda-
tions along the clinical management. Additional file 3
provides a checklist with the main recommendations
with a positive direction of action to assist in bedside
monitoring of clinical goals related to the recommenda-
tions and in the application of the management strategies.
Ventilatory support
1. We recommend using a lung-protective ventilation
strategy in all potential donors (low level of evidence,
strong recommendation).
Summary of evidence In potential donors, an initially
normal or near-normal lung function (PaO2/FiO2 300)
may deteriorate due to common complications in criti-
cal patients, such as pulmonary contusion, lung injury
following blood transfusion, pneumonia, atelectasis,
and mechanical ventilation-related iatrogenic injuries
[1318]. In addition, approximately 30–45% of poten-
tial donors develop acute respiratory distress syndrome
(ARDS; PaO2/FiO2 < 300), and only 15–20% of the lungs
are suitable for transplantation at the end of the procure-
ment process [13, 15, 17]. e lung-protective ventilation
strategy in potential donors with normal lungs and the
apnea testing performed with continuous positive airway
pressure (CPAP) have been associated with an increase in
eligibility for lung donation [1820].
Remarks e protective ventilation strategy for healthy
lungs consists of the combination of a tidal volume of
6–8 mL/kg and PEEP of 8–10-cm H2O. To promote
adequate blood oxygenation, FiO2 and PEEP must be
adjusted to obtain a SaO2 > 90%. To avoid atelectasis,
the apnea test with 10cm H2O CPAP can be performed
using a closed-circuit system in potential donors with
preserved lungs who are candidates for lung procure-
ment, or even when hypoxemic respiratory failure is
present. Also, the same procedure can be considered on
those who have failed the test due to hypoxemia after
disconnection.
2. We suggest not using alveolar recruitment maneu-
vers routinely in potential donors (very low level of
evidence, weak recommendation).
Summary of evidence Although alveolar recruitment
maneuvers have been suggested for the ventilatory
management of organ donors with lung injury (PaO2/
FiO2 < 300) [1316, 18, 20], and these maneuvers could
reduce hypoxemia after apnea testing, contributing to
increasing the viability of pulmonary grafts [1418, 20],
a randomized clinical trial showed unfavorable outcomes
in critically ill patients [21]. Besides, no randomized stud-
ies have demonstrated their efficacy in the population of
potential donors.
Remarks Performing alveolar recruitment maneuvers
in hemodynamically stable potential donors is probably
feasible in units with experience in the management of
ARDS. In cases of hypoxemia refractory to the lung-pro-
tective ventilation strategy, however, alveolar recruitment
maneuvers should not be performed routinely. eir use
is not indicated in hemodynamically unstable potential
donors.
Hemodynamic support
Volemic expansion andvasopressors
3. We recommend performing initial volemic expan-
sion in hemodynamically unstable potential donors
with hypovolemia or responsive to fluids according
to fluid responsiveness assessment (good clinical
practice).
4. We recommend administering norepinephrine or
dopamine to control blood pressure in potential
donors who remain hypotensive after volemic expan-
sion (very low level of evidence, strong recommenda-
tion).
Summary of evidence Potential donor hypotension is
associated with a higher incidence of postoperative liver
graft dysfunction and longer hospital stay in liver trans-
plant recipients [22, 23]. Targeting a mean arterial pres-
sure (MAP) 65mm Hg has also been associated with
reduced occurrence of cardiac arrest in potential donors
Page 4 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
Table 1 Summary ofrecommendations
Recommendations Level ofevidence Grade ofrecommendation Practical considerations
Ventilatory support
1. We recommend using a lung‑protective venti‑
lation strategy in all PDs
Low Strong Vt between 6 and 8 mL/kg of predicted body weight
and PEEP of 8–10‑cm H2O
Adjust FiO2 and PEEP to obtain SaO2 > 90%
Perform apnea testing with CPAP
2. We suggest not using ARM routinely in PDs Very low Weak ARM can be considered if there is refractory hypox‑
emia in hemodynamically stable PDs
Hemodynamic support
3. We recommend performing initial volemic
expansion in hemodynamically unstable PDs
with hypovolemia or responsive to fluids
according to fluid responsiveness assessment
Good clinical practice Initial volume expansion with 30 mL/kg of crystal‑
loids
Assess fluid status and responsiveness for additional
fluid replacement
Preferably use dynamic parameters
Neutral or negative fluid balance after achieving
hemodynamic stability
4. We recommend administering norepineph‑
rine or dopamine to control blood pressure
in PDs who remain hypotensive after volemic
expansion
Very low Strong Start adrenergic vasopressors to obtain a
MAP 65 mm Hg
Dopamine is the vasopressor of choice when there
is bradycardia
Consider the potential arrhythmogenic effect of
dopamine, which implies the risk of PD loss due to
cardiac arrest
5. We suggest not using low‑dose dopamine for
renal protection in PDs
Very low Weak Consider the potential arrhythmogenic effect of
dopamine, which implies the risk of PD loss due to
cardiac arrest
Endocrine and electrolyte management
6. We recommend combining AVP in PDs receiv‑
ing norepinephrine or dopamine
Low Strong Combine AVP (1 IU bolus + 0.5–2.4 IU/h) with norepi‑
nephrine or dopamine
7. We recommend administering AVP or DDAVP
to control polyuria in PDs with diabetes
insipidus
Low Strong AVP if vasopressors are required.
DDAVP (1–2‑µg IV 2–4 h) if vasopressors are not
required
8. We suggest combining low‑dose corticos‑
teroids in PDs receiving norepinephrine or
dopamine
Low Weak Combine 300 mg IV/day in PDs with norepinephrine
or dopamine
9. We suggest not using thyroid hormones
routinely in PDs
Very low Weak There are no hemodynamic benefits
They can be considered if prolonged management
is required
10. We suggest performing glycemic control in
PDs
Very low Weak Administer insulin to achieve a glucose level of
140–180 mg/dL
Monitor blood glucose at least every 6 h
11. We suggest maintaining serum sodium
levels < 155 mEq/dL in PDs
Very low Weak Correct water deficit with hypotonic fluids
Correct hypovolemia
12. We recommend maintaining serum potas‑
sium levels between 3.5 and 5.5 mEq/L in PDs
Very low Strong
13. We recommend maintaining serum magne‑
sium levels > 1.6 mEq/L in PDs
Very low Strong
Other aspects
14. We suggest maintaining nutritional support in
PDs if well tolerated
Very low Weak
15. We recommend using antibiotics in PDs with
infection or sepsis
Low Strong Maintain appropriate antibiotic therapy in the donor
for at least 24 h
Collect cultures from different sites in all donors
16. We suggest maintaining body temperature
above 35 °C in hemodynamically unstable PDs
Very low Weak Monitor core temperature
Prevent and treat hypothermia in PDs receiving
vasoactive amines
17. We suggest inducing hypothermia (34–35 °C)
in PDs without hemodynamic instability
Low Weak Monitor core temperature
Induce hypothermia by applying ice packs in PDs not
receiving vasoactive amines
18. We suggest transfusing packed red blood cells
in PDs with hemoglobin levels < 7 g/dL
Very low Weak
19. We suggest using goal‑directed protocols dur‑
ing the management of PDs
Very low Weak Monitor care using evidence‑based clinical goal‑
directed checklists
Page 5 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
[22, 24]. Intravascular volume expansion guided by ven-
tricular filling pressures or respiratory pulse pressure
variation (PPV) in hemodynamically unstable potential
donors is associated with faster recovery of renal graft
function and reduced circulating levels of inflammatory
cytokines [22, 25]. A randomized trial detected no differ-
ence between usual fluid management or fluid manage-
ment directed by a PPV and cardiac index. On the other
hand, there was a trend toward an increase in the number
of organs transplanted per donor among unstable poten-
tial donors responsive to fluids (p = 0.059) [26].
Conversely, avoiding fluid overload after the initial vol-
ume resuscitation to stabilize blood pressure seems to
be beneficial. is approach is associated with a greater
number of organs transplanted per donor and a greater
number of lungs transplanted without reducing the num-
ber of other donated organs or impairing survival in the
heart, liver, pancreas, or kidney transplant recipients [19,
2729].
If hypotension persists after adequate volume resusci-
tation, adrenergic vasopressors should be used to achieve
adequate blood-pressure levels [30]. ere is no dif-
ference in clinical outcomes in studies comparing nor-
epinephrine and dopamine [3133]. Disruption of vagal
activity secondary to brain death may result in atropine-
refractory bradycardia. In these cases, adrenergic drugs
Table 1 (continued)
PD: potential donor; Vt: total volume; PEEP: positive-end expiratory pressure; SaO2: ar terial oxygen saturation; CPAP: continuous positive airway pressure; ARM:
alveolar recruitment maneuver; MAP: mean arterial pressure; AVP: arginine-vasopressin; DDAVP: 1-deamino-8-d-arginine-vasopressin; IV: intravenous
Fig. 1 Flow of the recommendations
Page 6 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
as isoproterenol, epinephrine, and dopamine have been
suggested as positive chronotropic agents to treat brad-
ycardia in potential donors. Considering the predomi-
nance of noradrenaline action on alpha-1 receptors, its
infusion usually occurs without significant increase in
heart rate. Hence, dopamine or epinephrine may be more
convenient for the treatment of hypotension due to a
positive chronotropic effect [6, 34, 35].
Remarks Obtaining an MAP 65mm Hg as a blood-
pressure target contributes to the perfusion of organs
that are intended to be preserved for transplantation
[2224]. Hypovolemia is very frequent in potential organ
donors and should be considered when hypotension is
present. e initial infusion of crystalloids (e.g., 30mL/
kg) in potential donors who are hypovolemic or respon-
sive to fluids (when any fluid responsiveness assessment
parameter is already available) contributes to blood-pres-
sure control by improving tissue perfusion [2426].
Conversely, fluid overload should be avoided [19,
2729]. Assessment of fluid responsiveness with static
variables (e.g., central venous pressure—CVP) and/or
dynamic parameters (e.g., PPV) can be used to guide
volume replacement, helping to prevent fluid overload.
Dynamic parameters can more accurately discriminate
between responsive and unresponsive individuals [30
38]. Once hemodynamic stability is achieved, strategies
aimed at neutral fluid balance may be more beneficial
[19, 2729].
If the blood-pressure target is not achieved with the ini-
tial volume expansion, norepinephrine or dopamine infu-
sion should be started immediately. e use of dopamine
is likely advantageous for cases of bradycardia with signs
of low cardiac output [6, 34, 35], but the arrhythmogenic
potential of dopamine should be considered [39].
5. We suggest not using low-dose dopamine for renal
protection in potential donors (very low level of evi-
dence, weak recommendation).
Summary of evidence A cohort study of 93 heart trans-
plant recipients showed that pretreatment with low-dose
dopamine (4μg/kg/min) in heart donors was associated
with higher graft survival 3 years after transplantation
(87.0 vs. 67.8%, p < 0.03) [40]. A randomized-controlled
trial of 264 organ donors reported that the administra-
tion of low-dose dopamine reduced the need for hemodi-
alysis in recipients (OR 0.54; 95% CI 0.35–0.83), but with
no benefits for kidney graft survival after 3 years [41].
In the 5-year follow-up analysis of 487 renal transplant
recipients from the same trial, the researchers failed to
show a significant advantage of dopamine administration
in potential donors to long-term kidney graft survival,
although time of dopamine infusion and graft failure
were exposure-related (HR 0.96; 95% CI 0.92–1.00, per
hour) [42]. e same group reported that low-dose dopa-
mine did not negatively affect the short- or long-term
outcomes after liver transplants [43].
Remarks Although the administration of low-dose
dopamine in potential donors reduces the need for multi-
ple dialysis sessions, the long-term benefits for heart and
kidney graft survival are unclear. e panel considered
the potential arrhythmogenic effect of dopamine, which
may imply a greater risk of loss of potential donors due to
cardiac arrest before organ procurement.
Endocrine andelectrolyte management
Hormones
6. We recommend combining arginine vasopressin
(AVP) in potential donors receiving norepinephrine
or dopamine to control blood pressure (low level of
evidence, strong recommendation).
Summary of evidence e use of AVP in brain-dead
potential donors contributes to reducing the need for
adrenergic vasopressors and is associated with a lower
incidence of cardiovascular deterioration and cardiac
arrest [4448], in addition to contributing to the control
of plasma hyperosmolarity [46]. AVP infusion allows, in
some cases, complete discontinuation of adrenergic vaso-
pressors without causing adverse effects on the function
of organs transplanted [48, 49]. Finally, AVP infusion
seems to be associated with a greater number of donated
organs and a lower rate of graft refusal due to organ dys-
function [45].
Remarks e administration of an initial 1 IU AVP
bolus followed by infusion of 0.5IU/h to 2.4 UI/h helps
to maintain blood pressure in potential donors requiring
vasopressors, and contributes to the control of polyuria
and normovolemia in the presence of diabetes insipidus
[4446, 48, 49]. AVP should be started at the same time
of adrenergic vasopressor infusion.
7. We recommend administering AVP or 1-deam-
ino-8--arginine vasopressin (DDAVP) to control
polyuria in potential donors with diabetes insipidus
(low level of evidence, strong recommendation).
Summary of evidence e analysis of the database of a
randomized clinical trial that evaluated 487 renal graft
recipients showed better control of daily urine output
(p < 0.001) and a lower need for fluids in the DDAVP
group (p < 0.001). DDAVP was associated with improved
renal graft survival (85.4% vs. 73.6%, p = 0.003) af ter
2years, with no differences in acute rejections (OR 1.32;
Page 7 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
95% CI 0.70–2.49) or delayed graft function (OR 0.97;
95% CI 0.57–1.65) [50].
Remarks DDAVP acts exclusively on V2 receptors and
is indicated to control polyuria (urine output > 4 mL/
kg/h) in potential donors with diabetes insipidus who
maintain adequate blood pressure without adrenergic
vasopressors. AVP is preferred to control polyuria in
potential donors with diabetes insipidus who need adren-
ergic vasopressors. e combination of AVP and DDAVP
may be considered in refractory cases [51]. Although the
intranasal route is feasible, DDAVP should preferably be
administered intravenously, at a dose of 1–2µg every
2–4h [8, 13, 15], until a urine output < 4 mL/kg/h has
been achieved [5053].
8. We suggest using low-dose corticosteroids in poten-
tial donors receiving norepinephrine or dopamine to
control blood pressure (low level of evidence, weak
recommendation).
Summary of evidence A small retrospective study
reported that administration of 15-mg/kg methylpred-
nisolone was associated with higher PaO2/FiO2 values
(p = 0.01) and a greater number of lungs transplanted
(p < 0.01) [54]. Conversely, a before-and-after study
comparing 15-mg/kg methylprednisolone with 300-mg
hydrocortisone found no difference in the oxygenation
and hemodynamic stability of the potential donor or in
the number of organs transplanted [55]. A recent small
randomized-controlled trial showed that a single dose
of 15 mg/kg/day of methylprednisolone administered
to the potential organ donor may negatively affect the
graft function by increasing the antigenicity of the kid-
neys before transplantation. is negative effect was not
noticed among brain-dead donors who received 15mg/
kg/day of methylprednisolone followed by 100mg every
2 h until organ harvesting [56]. Eleven randomized-
controlled trials analyzed in a systematic review did not
support the use of high-dose corticosteroids in the man-
agement of potential donors [57]. On the other hand, a
randomized multicenter cluster study including 259
individuals compared the administration of low-dose
hydrocortisone (300 mg/day) with no corticosteroids.
e doses (p = 0.03) and duration of infusion (p < 0.001)
of vasopressors were lower in the intervention group, and
the complete vasopressor withdrawal was 4.7 times more
frequent in the corticosteroid group [58].
Remarks Despite conflicting evidence, the use of corti-
costeroids is of low cost and a low risk to potential donors
and may have a positive effect on hemodynamic out-
comes; therefore, their use is indicated in these patients.
Current evidence does not suggest ventilatory or hemo-
dynamic benefits associated with corticosteroid therapy
at high doses compared with low doses (i.e., 100mg every
8h). Higher doses should be avoided.
9. We suggest not using thyroid hormones routinely in
potential donors (very low level of evidence, weak
recommendation).
Summary of evidence Administration of thyroid hor-
mones in potential donors did not add any benefit, such
as a reduction in vasopressor use, an increase in cardiac
index, or an increase in organ procurement for trans-
plantation [5965]. Observational studies had suggested
an increase in heart procurement, which was not con-
firmed in randomized clinical trials [66, 67], even in
brain-dead organ donors with hemodynamic instability
and/or impaired cardiac function [68, 69].
Remarks Brain death is associated with a drop in circu-
lating thyroid hormone levels, which could contribute to
hemodynamic instability; however, there is no evidence
to support the use of thyroid hormones in potential
donors, given their costs and risks.
10. We suggest performing glycemic control in poten-
tial donors (very low level of evidence, weak rec-
ommendation).
Summary of evidence Four observational studies eval-
uated the effect of potential donor hyperglycemia on
post-transplant pancreatic function [7073]. One study
showed a correlation between donor blood glucose
immediately before organ retrieval and HbA1C 1 year
after transplantation [73], and another study found an
association between hyperglycemia and graft loss (HR
1.4; p = 0.03) [74]. Two studies showed no association
between potential donor blood glucose and post-trans-
plant pancreatic graft function [7072]. One obser-
vational study found an association between glycemic
control and creatinine of the potential donor before
organ retrieval [75]. Conversely, there is no evidence that
hyperglycemia is associated with liver graft dysfunction
[76]. A study of 1611 potential donors reported that a
glucose level < 180mg/dL was an independent predictor
of four or more organs transplanted per donor (OR 1.35;
95% CI 1.01–1.82) [77]. A set of potential donor care
measures, including glycemic control, was associated
with achieving 3 organs transplanted per donor (OR
1.9; 95% CI 1.35–2.68), but it was not possible to assess
the isolated effect of glycemic control [78].
Remarks Very-low-quality evidence suggests that a
glucose level < 180 mg/dL is associated with a greater
number of organs transplanted. Blood glucose should be
monitored in all potential donors at least every 6h, tar-
geting levels of 140–180mg/dL, and intravenous insulin
infusion can be used to this end.
Page 8 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
Electrolytes
11. We suggest maintaining serum sodium levels below
155mEq/dL in potential donors (very low level of
evidence, weak recommendation).
Summary of evidence. Five descriptive observational
studies were identified (n = 5733), which evaluated
only graft viability/function. In four of these studies
(n = 5545), there was no negative effect of donor hyper-
natremia above 155mEq/L on liver or heart graft func-
tion [7982]. In only one study (n = 188), hypernatremia
was associated with more cases of early graft loss [83].
Some authors have suggested that deceased-donor
hypernatremia may be a factor for worse prognosis of
graft function, but these findings have not been univer-
sally confirmed [7985]. Changes in natremia may reflect
inadequate volume management, especially in the pres-
ence of diabetes insipidus, one of the reasons for its cor-
rection [11].
Remarks Hypernatremia is often associated with hypo-
volemia, and should be controlled with volume expan-
sion, replacement of hypotonic fluids, and control of
polyuria with AVP or DDAVP. Serum sodium should be
monitored, targeting levels < 155mg/dL.
12. We recommend maintaining serum potassium lev-
els between 3.5 and 5.5mEq/L in potential donors
(very low level of evidence, strong recommenda-
tion).
Summary of evidence ere are no studies that directly
evaluate the effect of hyper- or hypokalemia in poten-
tial donors. A comparison of potassium levels in ICU
patients showed that hyperkalemia was more common
in patients who died (9.2% vs. 0.9%, p < 0.001) and that
serum potassium concentration could be a predictor of
death in critically ill patients [86].
Remarks Despite the absence of studies directly evalu-
ating the effects of potential donor serum potassium
levels, potassium is a determining factor in the resting
potential of electrically sensitive cells. Changes in potas-
sium levels are related to cardiac arrhythmias and may
compromise the management of potential donors. Potas-
sium levels should be monitored, and usual correction
measures should be implemented, targeting serum levels
between 3.5 and 5.5mEq/L.
13. We recommend maintaining serum magnesium
levels above 1.6 mEq/L in potential donors (very
low level of evidence, strong recommendation).
Summary of evidence Studies on the influence of serum
magnesium levels were found in critically ill patients,
but none in potential donors [8792]. Two observa-
tional studies and one randomized study identified
an association between hypomagnesemia and higher
mortality in critically ill patients [87, 88, 91], in addi-
tion to a greater likelihood of QT interval prolongation
(OR 42.8; 95% CI 14.5–126.2) [88]. is association of
hypomagnesemia with mortality was reinforced in a sys-
tematic review [89]. In addition to being arrhythmogenic,
hypomagnesemia appears to be associated with non-
recovery of renal function in patients with acute kidney
injury (70% vs. 31%, p = 0.003) [92].
Remarks Hypomagnesemia is associated with cardiac
arrhythmias and worse prognosis in critically ill patients,
with no direct evidence in brain-dead potential donors.
However, this is a low-cost procedure, and in the ICU
setting, routine monitoring until normalization of mag-
nesium levels is a common practice, which may be ben-
eficial for potential donors. Magnesium levels should be
monitored, and magnesium sulfate should be adminis-
tered, as usual, targeting serum levels above 1.6mEq/L.
Other aspects ofpotential donor management
Nutritional support
14. We suggest maintaining nutritional support in
potential donors if well tolerated (very low level of
evidence, weak recommendation).
Summary of evidence Although there is no evidence on
nutritional support, different guidelines recommend con-
tinuing nutritional support of the donor in the absence
of contraindications [7, 9, 51]. Possible benefits include
increased liver glycogen reserves, which could posi-
tively influence the liver graft [93, 94], and maintenance
of intestinal mucosal trophism, which could reduce the
potential for bacterial translocation.
Remarks For brain-dead individuals requiring ICU
management for prolonged periods (e.g., brain-dead
pregnant women; prolongation of the diagnostic pro-
cess or the family decision for donation), it is reasonable
that energy expenditure should be estimated or meas-
ured [95], considering that baseline energy expenditure
is 15–30% lower in brain-dead individuals than in other
critically ill patients [96]. us, in individuals already
receiving full nutritional support, energy intake may be
reduced once brain death is established. A minimum
energy intake (e.g., 500 kcal) could be considered in
potential donors who had not been on enteral feeding
before brain death was diagnosed, taking into account its
potential benefit in the maintenance of intestinal mucosal
trophism. However, it does not seem appropriate to start
enteral feeding when the organs are likely to be har-
vested within a short period or in the presence of any of
the usual contraindications to initiate/maintain enteral
feeding (e.g., gastrointestinal tract obstruction, ileus,
Page 9 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
vomiting/aspiration of gastric contents, severe hemody-
namic instability, and high doses of vasopressors).
Infection andsepsis
15. We recommend using antibiotics in potential
donors with infection or sepsis (low level of evi-
dence, strong recommendation).
Summary of evidence Different observational studies
evaluated the transmission of bacterial infection in organ
donors with culture-proven infection. e most com-
monly observed microorganisms were Staphylococcus
aureus, Streptococcus sp., Klebsiella sp., and Acinetobac-
ter baumannii. Bacterial transmission is rarely observed
[97104], provided that donors with evidence of infec-
tion receive appropriate antibiotic therapy [97102, 105,
106]. e duration of donor antibiotic therapy ranged
from 24 to 96h in different studies [97, 99, 102, 105].
Also, different authors have reported maintaining the
same antibiotics administered to the donors in the trans-
plant recipients, for periods ranging from 7 to 14days
[98, 100, 105, 106]. e presence of donor infection had
no impact on the survival of grafts or transplant recipi-
ents [97102, 105, 106].
Remarks e risk of transmission of bacterial infection
from organ donors to recipients is low, and donor infec-
tion does not appear to negatively affect outcomes. e
risks are lower with appropriate antibiotic therapy in the
donor for at least 24h, followed by maintenance of the
antibiotic in the recipient for 7–14days [97102, 105,
106]. Some donors have subclinical bacteremia at the
time of organ procurement; therefore, cultures should be
collected from blood and different sites in all donors, and
the recipient antibiotic therapy should be directed by the
results of culture [99, 107110].
Body temperature control
16. We suggest maintaining body temperature above
35 °C in hemodynamically unstable potential
donors (very low level of evidence, weak recom-
mendation).
17. We suggest inducing moderate hypothermia (34–
35°C) in potential donors without hemodynamic
instability (low level of evidence, weak recommen-
dation).
Summary of evidence Delayed renal graft function
was evaluated in a randomized-controlled trial that
compared hypothermia (34–35°C) versus usual man-
agement (36.5–37.5°C) in 370 potential donors without
hemodynamic instability. e main result was a reduc-
tion in delayed renal graft function among recipients
(OR 0.62; 95% CI 0.43–0.92). ere was no differ-
ence in the number of organs transplanted per donor,
adverse events, or cardiac arrest [111]. Two retrospec-
tive cohort studies nested in the randomized dopamine
trial demonstrated that spontaneous donor hypother-
mia was associated with lower creatinine levels before
organ procurement without effect on kidney graft
survival [112], and with an unfavorable clinical course
after heart transplant [113]. In a clinical population of
post-cardiac arrest patients, i.e., patients at increased
risk of hemodynamic instability, a meta-analysis of five
clinical trials found a higher risk of recurrent arrest in
patients with induced hypothermia (< 35°C) in prehos-
pital management (RR 1.23; 95% CI 1.02–1.48) [114].
Remarks Hypothermia is a low-cost intervention
[115] associated with better renal graft function, but it
can increase the risk of cardiac arrest in the potential
donor [111, 114]. e risk appears to be low in hemo-
dynamically stable potential donors, in whom the use
of hypothermia can be justified by improved graft via-
bility. In the presence of hemodynamic instability [111],
normothermia (> 35°C) should be maintained in poten-
tial donors to reduce the risk of cardiac arrest [114].
Induction of moderate hypothermia (34–35°C) is con-
sidered a simple (application of ice packs) and inex-
pensive approach, but it is important to monitor core
temperature, which is not available in all ICUs.
Red blood cell transfusion
18. We suggest transfusing packed red blood cells in
potential donors with hemoglobin levels < 7 g/dL
(very low level of evidence, weak recommenda-
tion).
Summary of evidence e systematic literature search
identified 1 descriptive observational study that evalu-
ated function in 1884 renal grafts from 1006 brain-dead
donors. Among donors, 52% received blood transfu-
sion. Renal grafts from transfused donors had a lower
rate of delayed graft function than those from non-
transfused donors (26% vs. 34%, p < 0.001). e criteria
defining the need for blood transfusion were not identi-
fied [116].
Remarks Anemia can compromise oxygen transport
and delivery to the organs that are intended to be pre-
served for transplantation. However, we are unaware of
the hemoglobin concentration necessary to contribute
to adequate oxygen transport and delivery in potential
donors. Considering the high cost and frequent shortage
of blood products for transfusion, the decision to trans-
fuse should not differ from the usual practice in other
critically ill patients.
Page 10 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
Goal‑directed protocols
19. We suggest using a goal-directed protocol during
the management of potential donors (very low level
of evidence, weak recommendation).
Summary of evidence Although there is no consistent
evidence about an individual treatment that will improve
the number and quality of donated organs [117], obser-
vational studies have reported that combining different
treatments through the application of a potential donor-
management protocol is associated with a higher organ
yield for transplantation [24, 78, 118124], lower inci-
dence of delayed renal graft function [111], greater eligi-
bility for lung donation [19, 28], and lower incidence of
donor losses due to cardiac arrest [19, 24, 28, 119, 120].
In general, the outcomes are associated with the number
of goals achieved during potential donor management,
including ventilatory, hemodynamic, and endocrine-
metabolic management goals [24, 78, 121123]. In seven
studies, the use of a checklist helped implement the goal-
directed protocols and may have positively influenced the
results [19, 28, 78, 121, 124126].
Remarks e application of a potential donor-manage-
ment protocol guided by a clinical goal-directed checklist
may contribute to increasing the number of organs trans-
planted per donor, influence graft function, and reduce
donor losses due to cardiac arrest.
General considerations andfuture directions
e present guideline aimed to provide parameters to
optimize the clinical management of potential donors
based on the available evidence, aiming to improve the
quality of organs for transplantation and to reduce donor
losses. However, it is well known that it may take years
for a large-scale translation of the best scientific evidence
into effective practice. us, establishing clinical proto-
cols can help to reduce the time required to incorporate
best practices. e use of a goal-directed checklist can
play an important role in the adjustment of approaches
and adherence to the best evidence in complex proce-
dures [127130].
is guideline evaluated a broad volume of treatments
and we performed rigorous PICO-driven research to
provide the recommendations based on standardized
rapid review methods [9, 10]. Potential limitations are
the low or very low certainty in the evidence identified
for many of the questions, and indirect evidence that did
not change after the systematic review update. However,
management recommendations are consistent with simi-
lar documents recently published [11, 131, 132].
Several challenges regarding ethical, infrastructure, and
operational issues are faced while planning and conduct-
ing studies that involve potential organ donors, which
results in few randomized clinical trials [133]. e scar-
city of studies with such methodological strength implies
uncertainties about some interventions such as low-dose
dopamine and moderate hypothermia, which, despite
appearing to be related to renal graft benefit, may result
in cardiac arrhythmias and hemodynamic instability.
In this context, developing clinical trials in this field of
medical knowledge may be helpful to understand some
important aspects in the management of the potential
organ donor.
Some observational studies have reported that the
application of a checklist to guide the management of
brain-dead potential donors may help to reduce the rate
of cardiac arrest in potential donors and increase the
number of organs recovered per donor [24, 78, 119, 121,
122, 124, 126, 134, 135]. In this context, we used the main
recommendations of the present guideline to develop an
evidence-based clinical goal-directed checklist (Addi-
tional file 3) with the purpose of providing transplant
coordinators and ICU professionals with essential infor-
mation to optimize the care of potential donors.
However, because the available studies highlighting
the role of potential donor-management checklists are
observational, there is insufficient evidence to support
the systematic use of checklists in the management of
potential donors. erefore, we proposed the Donation
Network to Optimize Organ Recovery Study (DONORS;
NCT03179020), which is a parallel cluster randomized-
controlled multicenter trial that aims to test the effec-
tiveness of the implementation of a checklist containing
goals and recommendations of care in reducing organ
donor losses due to cardiac arrest and increasing the
number of organs recovered per donor [136].
e implementation of the checklist should be pre-
ceded by the appropriate training of intensive care teams
and transplant coordinators. We suggest applying the
checklist at the bedside immediately after the first clinical
examination for the diagnosis of brain death, repeating
the application, ideally, every 6 h until organ procure-
ment for transplantation. We also suggest that a mem-
ber of the transplant coordination office or a designated
professional of the ICU or emergency department applies
the checklist at the bedside. e same individual will also
be responsible for personally prompting the physician in
charge to modify the clinical management if any inappro-
priate aspect of care, according to the checklist, is noted.
Page 11 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
Supplementary information
Supplementary information accompanies this paper at https ://doi.
org/10.1186/s1361 3‑020‑00787 ‑0.
Additional le1. Working group and contributions of each participant.
Additional le2. Declaration of competing interests.
Additional le3. Checklist for clinical management of brain‑dead poten‑
tial organ donor.
Acknowledgements
The authors would like to thank the Brazilian Ministry of Health and the Gen‑
eral Coordination Office of the National Transplant System (CGSNT), as well as
Moinhos de Vento Hospital, the Brazilian Association of Organ Transplantation
(ABTO), the Brazilian Association of Intensive Care Medicine (AMIB) Committee
for Organ Donation for Transplant, and the Brazilian Research in Intensive Care
Network (BRICNet) for their support.
Authors’ contributions
All authors, except for AR, DFSP, FDP, RCM, and RRN, participated in at least one
of the expert panels. All authors read and approved the final manuscript. The
detailed contribution of each author is presented in Additional file 1.
Funding
This guideline was funded by the Brazilian Ministry of Health through the
Programa de Apoio ao Desenvolvimento Institucional do Sistema Único de
Saúde (PROADI‑SUS). The funding body has no role in the coordination of the
guideline.
Availability of data and materials
All relevant data are within the paper and its additional files.
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The competing interests of each author are presented in Additional file 2.
Author details
1 Hospital Moinhos de Vento (HMV), R. Ramiro Barcelos, 910, Porto Alegre,
RS 90035000, Brazil. 2 Hospital Municipal São José (HMSJ), Joinville, SC, Brazil.
3 Centro Hospitalar Unimed, Joinville, SC, Brazil. 4 Hospital do Coração (HCor),
R. Desembargador Eliseu Guilherme, 147, São Paulo, SP 04004030, Brazil. 5 Uni‑
versidade da Região de Joinville (UNIVILLE), R. Paulo Malschitzki, 10, Joinville,
SC 89219710, Brazil. 6 Clínica de Nefrologia de Joinville, R. Plácido Gomes, 370,
Joinville, SC 89202‑050, Brazil. 7 Hospital de Clínicas de Porto Alegre (HCPA),
R. Ramiro Barcelos, 2350, Porto Alegre, RS 90035007, Brazil. 8 Universidade
Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Sarmento Leite, 245,
Porto Alegre, RS 90050‑170, Brazil. 9 Hospital de Pronto de Socorro (HPS), Porto
Alegre, RS, Brazil. 10 General Coordination Office of the National Transplant
System, Brazilian Ministry of Health, Esplanada dos Ministérios, Bloco G, Edifício
Sede, Brasília, DF 70058900, Brazil. 11 Faculdade de Medicina, Universidade
de São Paulo (USP), Av. Dr, Arnaldo 455, Sala 3206, São Paulo, SP 01246903,
Brazil. 12 Universidade do Extremo Sul Catarinense (UNESC), Av. Universitária,
1105, Criciúma, SC 88806000, Brazil. 13 Intensive Care Unit, Hospital São José,
R. Cel. Pedro Benedet, 630, Criciúma, SC 88801‑250, Brazil. 14 National Institute
of Infectious Disease Evandro Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Av.
Brasil, 4365, Rio de Janeiro, RJ 21040360, Brazil. 15 Instituto D’Or de Pesquisa
e Ensino (IDOR), R. Diniz Cordeiro, 30, Rio de Janeiro, RJ 22281100, Brazil.
16 Hospital São Paulo (HU), Universidade Federal de São Paulo (UNIFESP), R.
Napoleão de Barros 737, São Paulo, SP 04024002, Brazil. 17 Organização de
Procura de Órgãos e Tecidos de Santa Catarina (OPO/SC), Rua Esteves Júnior,
390, Florianópolis, SC 88015130, Brazil. 18 Hospital Sírio‑Libanês, R. Dona Adma
Jafet, 115, São Paulo, SP, Brazil. 19 Universidade Federal do Rio Grande do Sul
(UFRGS), Ramiro Barcelos, 2350, Porto Alegre, RS 90035007, Brazil. 20 Facul‑
dade de Medicina de São José do Rio Preto, Av Faria Lima, 5544, São José
do Rio Preto, SP 15090000, Brazil. 21 National Institute for Health Technology
Assessment, UFRGS, Rua Ramiro Barcelos, 2350, Porto Alegre, RS 90035903,
Brazil. 22 Department of Health Research Methods, Evidence, and Impact (HEI),
McMaster University, 1280 Main St W, Hamilton, ON, Canada.
Received: 3 August 2020 Accepted: 1 December 2020
References
1. Tullius SG, Rabb H. Improving the supply and quality of deceased‑
donor organs for transplantation. N Engl J Med. 2018;378:1920–9. https
://doi.org/10.1056/NEJMr a1507 080.
2. The Madrid resolution on organ donation and transplantation. national
responsibility in meeting the needs of patients, guided by the WHO
principles. Transplantation. 2011;91(Suppl 11):S29–31. https ://doi.
org/10.1097/01.tp.00003 99131 .74618 .a5.
3. Dominguez‑Gil B, Delmonico FL, Shaheen FA, Matesanz R, O’Connor K,
Minina M, et al. The critical pathway for deceased donation: report‑
able uniformity in the approach to deceased donation. Transpl Int.
2011;24:373–8. https ://doi.org/10.1111/j.1432‑2277.2011.01243 .x.
4. DuBose J, Salim A. Aggressive organ donor management protocol. J
Intensive Care Med. 2008;23:367–75. https ://doi.org/10.1177/08850
66608 32420 8.
5. Powner D. Aggressive donor care–to what end? J Intensive Care Med.
2008;23:409–11. https ://doi.org/10.1177/08850 66608 32419 8.
6. Westphal GA, Caldeira Filho M, Vieira KD, Zaclikevis VR, Bartz MC,
Wanzuita R, et al. Guidelines for potential multiple organ donors (adult):
part I. Overview and hemodynamic support. Rev Bras Ter Intensiva.
2011;23:255–68.
7. Westphal GA, Caldeira Filho M, Vieira KD, Zaclikevis VR, Bartz MC, Wan‑
zuita R, et al. Guidelines for potential multiple organ donors (adult): part
II. Mechanical ventilation, endocrine metabolic management, hemato‑
logical and infectious aspects. Rev Bras Ter Intensiva. 2011;23:269–82.
8. Westphal GA, Caldeira Filho M, Vieira KD, Zaclikevis VR, Bartz MC,
Wanzuita R, et al. Guidelines for potential multiple organ donors
(adult). Part III: organ‑specific recommendations. Rev Bras Ter Intensiva.
2011;23:410–25.
9. Schunemann HJ, Moja L. Reviews: rapid! Rapid! Rapid!… and system‑
atic. Syst Rev. 2015;4:4. https ://doi.org/10.1186/2046‑4053‑4‑4.
10. Haby MM, Chapman E, Clark R, Barreto J, Reveiz L, Lavis JN. Designing
a rapid response program to support evidence‑informed decision‑
making in the Americas region: using the best available evidence and
case studies. Implement Sci. 2016;11:117. https ://doi.org/10.1186/s1301
2‑016‑0472‑9.
11. Kotloff RM, Blosser S, Fulda GJ, Malinoski D, Ahya VN, Angel L, et al.
Management of the potential organ donor in the ICU: Society of Critical
Care Medicine/American College of Chest Physicians/Association of
Organ Procurement Organizations Consensus Statement. Crit Care
Med. 2015;43:1291–325. https ://doi.org/10.1097/CCM.00000 00000
00095 8.
12. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck‑Ytter Y, Alonso‑Coello P,
et al. GRADE: an emerging consensus on rating quality of evidence
and strength of recommendations. BMJ. 2008;336:924–6. https ://doi.
org/10.1136/bmj.39489 .47034 7.AD.
13. Mascia L, Zavala E, Bosma K, Pasero D, Decaroli D, Andrews P, et al. High
tidal volume is associated with the development of acute lung injury
after severe brain injury: an international observational study. Crit Care
Med. 2007;35:1815–20. https ://doi.org/10.1097/01.CCM.00002 75269
.77467 .DF.
14. Mascia L, Pasero D, Slutsky AS, Arguis MJ, Berardino M, Grasso S, et al.
Effect of a lung protective strategy for organ donors on eligibility and
availability of lungs for transplantation: a randomized controlled trial.
JAMA. 2010;304:2620–7. https ://doi.org/10.1001/jama.2010.1796.
15. Lebovitz DJ, Reis K, Yun J, Herman L, McCurry KR. An aggressive lung
recruitment protocol increases the percentage of lung donors with
no increased adverse effect in lung recipients: 3173. Transplantation.
2010;90:356.
Page 12 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
16. Noiseux N, Nguyen BK, Marsolais P, Dupont J, Simard L, Houde I, et al.
Pulmonary recruitment protocol for organ donors: a new strategy to
improve the rate of lung utilization. Transplant Proc. 2009;41:3284–9.
https ://doi.org/10.1016/j.trans proce ed.2009.08.041.
17. Gabbay E, Williams TJ, Griffiths AP, Macfarlane LM, Kotsimbos TC, Esmore
DS, et al. Maximizing the utilization of donor organs offered for lung
transplantation. Am J Respir Crit Care Med. 1999;160:265–71. https ://
doi.org/10.1164/ajrcc m.160.1.98110 17.
18. Gattinoni L, Carlesso E, Brazzi L, Caironi P. Positive end‑expiratory
pressure. Curr Opin Crit Care. 2010;16:39–44. https ://doi.org/10.1097/
MCC.0b013 e3283 35472 3.
19. Minambres E, Coll E, Duerto J, Suberviola B, Mons R, Cifrian JM, et al.
Effect of an intensive lung donor‑management protocol on lung trans‑
plantation outcomes. J Heart Lung Transplant. 2014;33:178–84. https ://
doi.org/10.1016/j.healu n.2013.10.034.
20. Angel LF, Levine DJ, Restrepo MI, Johnson S, Sako E, Carpenter A, et al.
Impact of a lung transplantation donor‑management protocol on
lung donation and recipient outcomes. Am J Respir Crit Care Med.
2006;174:710–6. https ://doi.org/10.1164/rccm.20060 3‑432OC .
21. Writing Group for the Alveolar Recruitment for Acute Respiratory
Distress Syndrome Trial I, Cavalcanti AB, Suzumura EA, Laranjeira LN,
Paisani DM, Damiani LP, et al. Effect of lung recruitment and Titrated
Positive End‑Expiratory Pressure (PEEP) vs Low PEEP on mortality in
patients with acute respiratory distress syndrome: a randomized clinical
trial. JAMA. 2017;318:1335–45. https ://doi.org/10.1001/jama.2017.14171
.
22. Gruenberger T, Steininger R, Sautner T, Mittlbock M, Muhlbacher F. Influ‑
ence of donor criteria on postoperative graft function after orthotopic
liver transplantation. Transpl Int. 1994;7(Suppl 1):S672–4. https ://doi.
org/10.1111/j.1432‑2277.1994.tb014 70.x.
23. delaTorre AN, Kuo PC, Plotkin JS, Ridge LA, Howell CD, Bartlett ST, et al.
Influence of donor base deficit status on recipient outcomes in liver
transplantation. Transplant Proc. 1997;29:474. https ://doi.org/10.1016/
s0041 ‑1345(96)00627 ‑6.
24. Westphal GA, Coll E, deSouza RL, Wagner S, Montemezzo A, Cani
de Souza FC, et al. Positive impact of a clinical goal‑directed proto‑
col on reducing cardiac arrests during potential brain‑dead donor
maintenance. Crit Care. 2016;20:323. https ://doi.org/10.1186/s1305
4‑016‑1484‑1.
25. Murugan R, Venkataraman R, Wahed AS, Elder M, Carter M, Madden NJ,
et al. Preload responsiveness is associated with increased interleu‑
kin‑6 and lower organ yield from brain‑dead donors. Crit Care Med.
2009;37:2387–93. https ://doi.org/10.1097/CCM.0b013 e3181 a960d 6.
26. Al‑Khafaji A, Elder M, Lebovitz DJ, Murugan R, Souter M, Stuart S, et al.
Protocolized fluid therapy in brain‑dead donors: the multicenter rand‑
omized MOnIToR trial. Intensive Care Med. 2015;41:418–26. https ://doi.
org/10.1007/s0013 4‑014‑3621‑0.
27. Abdelnour T, Rieke S. Relationship of hormonal resuscitation therapy
and central venous pressure on increasing organs for transplant. J
Heart Lung Transplant. 2009;28:480–5. https ://doi.org/10.1016/j.healu
n.2009.01.018.
28. Minambres E, Perez‑Villares JM, Chico‑Fernandez M, Zabalegui A,
Duenas‑Jurado JM, Misis M, et al. Lung donor treatment protocol
in brain dead‑donors: a multicenter study. J Heart Lung Transplant.
2015;34:773–80. https ://doi.org/10.1016/j.healu n.2014.09.024.
29. Minambres E, Perez‑Villares JM, Terceros‑Almanza L, Duenas‑Jurado JM,
Zabalegui A, Misis M, et al. An intensive lung donor treatment protocol
does not have negative influence on other grafts: a multicentre study.
Eur J Cardiothorac Surg. 2016;49:1719–24. https ://doi.org/10.1093/ejcts
/ezv45 4.
30. Cecconi M, De Backer D, Antonelli M, Beale R, Bakker J, Hofer C, et al.
Consensus on circulatory shock and hemodynamic monitoring. Task
force of the European Society of Intensive Care Medicine. Intensive Care
Med. 2014;40:1795–815. https ://doi.org/10.1007/s0013 4‑014‑3525‑z.
31. Schnuelle P, Lorenz D, Mueller A, Trede M, Van Der Woude FJ. Donor
catecholamine use reduces acute allograft rejection and improves graft
survival after cadaveric renal transplantation. Kidney Int. 1999;56:738–
46. https ://doi.org/10.1046/j.1523‑1755.1999.00567 .x.
32. Schnuelle P, Berger S, de Boer J, Persijn G, van der Woude FJ. Effects of
catecholamine application to brain‑dead donors on graft survival in
solid organ transplantation. Transplantation. 2001;72:455–63. https ://
doi.org/10.1097/00007 890‑20010 8150‑00017 .
33. von Ziegler F, Helbig S, Kreissl N, Meiser B, Becker A, Kaczmarek I.
Norepinephrine versus dopamine pretreatment of potential heart
donors‑impact on long‑term outcome. Ann Transplant. 2013;18:320–6.
https ://doi.org/10.12659 /AOT.88396 0.
34. Dictus C, Vienenkoetter B, Esmaeilzadeh M, Unterberg A, Ahmadi
R. Critical care management of potential organ donors: our current
standard. Clin Transplant. 2009;23(Suppl 21):2–9. https ://doi.org/10.1
111/j.1399‑0012.2009.01102 .x.
35. Wood KE, Becker BN, McCartney JG, D’Alessandro AM, Coursin DB.
Care of the potential organ donor. N Engl J Med. 2004;351(26):2730–
9. https ://doi.org/10.1056/NEJMr a0131 03.
36. Marik PE, Baram M, Vahid B. Does central venous pressure predict
fluid responsiveness? A systematic review of the literature and the
tale of seven mares. Chest. 2008;134:172–8. https ://doi.org/10.1378/
chest .07‑2331.
37. Eskesen TG, Wetterslev M, Perner A. Systematic review including
re‑analyses of 1148 individual data sets of central venous pressure as
a predictor of fluid responsiveness. Intensive Care Med. 2016;42:324–
32. https ://doi.org/10.1007/s0013 4‑015‑4168‑4.
38. Marik PE, Cavallazzi R, Vasu T, Hirani A. Dynamic changes in arterial
waveform derived variables and fluid responsiveness in mechani
cally ventilated patients: a systematic review of the literature. Crit
Care Med. 2009;37:2642–7. https ://doi.org/10.1097/CCM.0b013 e3181
a590d a.
39. Rui Q, Jiang Y, Chen M, Zhang N, Yang H, Zhou Y. Dopamine versus
norepinephrine in the treatment of cardiogenic shock: a PRISMA‑
compliant meta‑analysis. Medicine. 2017;96:e8402. https ://doi.
org/10.1097/MD.00000 00000 00840 2.
40. Benck U, Hoeger S, Brinkkoetter PT, Gottmann U, Doenmez D,
Boesebeck D, et al. Effects of donor pre‑treatment with dopamine
on survival after heart transplantation: a cohort study of heart
transplant recipients nested in a randomized controlled multicenter
trial. J Am Coll Cardiol. 2011;58:1768–77. https ://doi.org/10.1016/j.
jacc.2011.05.060.
41. Schnuelle P, Gottmann U, Hoeger S, Boesebeck D, Lauchart W, Weiss C,
et al. Effects of donor pretreatment with dopamine on graft function
after kidney transplantation: a randomized controlled trial. JAMA.
2009;302:1067–75. https ://doi.org/10.1001/jama.2009.1310.
42. Schnuelle P, Schmitt WH, Weiss C, Habicht A, Renders L, Zeier M,
et al. Effects of dopamine donor pretreatment on graft survival after
kidney transplantation: a randomized trial. Clin J Am Soc Nephrol.
2017;12:493–501. https ://doi.org/10.2215/CJN.07600 716.
43. Benck U, Jung M, Kruger B, Grimm A, Weiss C, Yard BA, et al. Donor
Dopamine Does Not Affect Liver Graft Survival: evidence of Safety From
a Randomized Controlled Trial. Liver Transpl. 2018;24:1336–45. https ://
doi.org/10.1002/lt.25301 .
44. Iwai A, Sakano T, Uenishi M, Sugimoto H, Yoshioka T, Sugimoto T. Effects
of vasopressin and catecholamines on the maintenance of circulatory
stability in brain‑dead patients. Transplantation. 1989;48:613–7.
45. Kinoshita Y, Yahata K, Yoshioka T, Onishi S, Sugimoto T. Long‑term renal
preservation after brain death maintained with vasopressin and epi‑
nephrine. Transpl Int. 1990;3:15–8. https ://doi.org/10.1007/bf003 33196 .
46. Pennefather SH, Bullock RE, Mantle D, Dark JH. Use of low dose arginine
vasopressin to support brain‑dead organ donors. Transplantation.
1995;59:58–62. https ://doi.org/10.1097/00007 890‑19950 1150‑00011 .
47. Plurad DS, Bricker S, Neville A, Bongard F, Putnam B. Arginine vasopres‑
sin significantly increases the rate of successful organ procurement in
potential donors. Am J Surg. 2012;204:856–60. https ://doi.org/10.1016/j.
amjsu rg.2012.05.011.
48. Chen JM, Cullinane S, Spanier TB, Artrip JH, John R, Edwards NM, et al.
Vasopressin deficiency and pressor hypersensitivity in hemodynami‑
cally unstable organ donors. Circulation. 1999;100:244–6. https ://doi.
org/10.1161/01.cir.100.suppl _2.ii‑244.
49. K atz K, Lawler J, Wax J, O’Connor R, Nadkarni V. Vasopressin pressor
effects in critically ill children during evaluation for brain death and
organ recovery. Resuscitation. 2000;47:33–40. https ://doi.org/10.1016/
s0300 ‑9572(00)00196 ‑9.
50. Benck U, Gottmann U, Hoeger S, Lammert A, Rose D, Boesebeck D, et al.
Donor desmopressin is associated with superior graft survival after
Page 13 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
kidney transplantation. Transplantation. 2011;92:1252–8. https ://doi.
org/10.1097/TP.0b013 e3182 36cd4 c.
51. Shemie SD, Ross H, Pagliarello J, Baker AJ, Greig PD, Brand T, et al.
Organ donor management in Canada: recommendations of the forum
on Medical Management to Optimize Donor Organ Potential. CMAJ.
2006;174:S13–32. https ://doi.org/10.1503/cmaj.04513 1.
52. Gramm HJ, Meinhold H, Bickel U, Zimmermann J, von Hammerstein B,
Keller F, et al. Acute endocrine failure after brain death? Transplantation.
1992;54:851–7. https ://doi.org/10.1097/00007 890‑19921 1000‑00016 .
53. Fiser DH, Jimenez JF, Wrape V, Woody R. Diabetes insipidus in
children with brain death. Crit Care Med. 1987;15:551–3. https ://doi.
org/10.1097/00003 246‑19870 6000‑00002 .
54. Follette DM, Rudich SM, Babcock WD. Improved oxygenation and
increased lung donor recovery with high‑dose steroid administration
after brain death. J Heart Lung Transplant. 1998;17:423–9. https ://doi.
org/10.1007/s8010 90000 086.
55. Dhar R, Cotton C, Coleman J, Brockmeier D, Kappel D, Marklin G,
et al. Comparison of high‑ and low‑dose corticosteroid regimens for
organ donor management. J Crit Care. 2013;28(111):e1–7. https ://doi.
org/10.1007/s8010 90000 086.
56. Jafari R, Aflatoonian R, Falak R, Pourmand G, Dehghani S, Mortazavi M,
et al. Down‑regulation of inflammatory signaling pathways despite
up‑regulation of Toll‑like receptors; the effects of corticosteroid therapy
in brain‑dead kidney donors, a double‑blind, randomized, controlled
trial. Mol Immunol. 2018;94:36–44. https ://doi.org/10.1016/j.molim
m.2017.12.012.
57. Dupuis S, Amiel JA, Desgroseilliers M, Williamson DR, Thiboutot Z, Serri
K, et al. Corticosteroids in the management of brain‑dead potential
organ donors: a systematic review. Br J Anaesth. 2014;113:346–59. https
://doi.org/10.1093/bja/aeu15 4.
58. Pinsard M, Ragot S, Mertes PM, Bleichner JP, Zitouni S, Cook F, et al.
Interest of low‑dose hydrocortisone therapy during brain‑dead organ
donor resuscitation: the CORTICOME study. Crit Care. 2014;18:R158.
https ://doi.org/10.1186/cc139 97.
59. Venkateswaran RV, Steeds RP, Quinn DW, Nightingale P, Wilson IC,
Mascaro JG, et al. The haemodynamic effects of adjunctive hormone
therapy in potential heart donors: a prospective randomized double‑
blind factorially designed controlled trial. Eur Heart J. 2009;30:1771–80.
https ://doi.org/10.1093/eurhe artj/ehp08 6.
60. Perez‑Blanco A, Caturla‑Such J, Canovas‑Robles J, Sanchez‑Paya J.
Efficiency of triiodothyronine treatment on organ donor hemodynamic
management and adenine nucleotide concentration. Intensive Care
Med. 2005;31:943–8. https ://doi.org/10.1007/s0013 4‑005‑2662‑9.
61. Jeevanandam V. Triiodothyronine: spectrum of use in heart transplanta‑
tion. Thyroid. 1997;7:139–45. https ://doi.org/10.1089/thy.1997.7.139.
62. Goarin JP, Cohen S, Riou B, Jacquens Y, Guesde R, Le Bret F, et al. The
effects of triiodothyronine on hemodynamic status and cardiac func‑
tion in potential heart donors. Anesth Analg. 1996;83:41–7. https ://doi.
org/10.1097/00000 539‑19960 7000‑00008 .
63. Randell TT, Hockerstedt KA. Triiodothyronine treatment in brain‑dead
multiorgan donors–a controlled study. Transplantation. 1992;54:736–8.
https ://doi.org/10.1097/00007 890‑19921 0000‑00034 .
64. Garcia‑Fages LC, Antolin M, Cabrer C, Talbot R, Alcaraz A, Lozano F, et al.
Effects of substitutive triiodothyronine therapy on intracellular nucleo‑
tide levels in donor organs. Transplant Proc. 1991;23:2495–6.
65. Mariot J, Jacob F, Voltz C, Perrier JF, Strub P. Value of hormonal treatment
with triiodothyronine and cortisone in brain dead patients. Ann Fr
Anesth Reanim. 1991;10:321–8. https ://doi.org/10.1007/s8010 90000
086.
66. Macdonald PS, Aneman A, Bhonagiri D, Jones D, O’Callaghan G, Sil‑
vester W, et al. A systematic review and meta‑analysis of clinical trials of
thyroid hormone administration to brain dead potential organ donors.
Crit Care Med. 2012;40:1635–44. https ://doi.org/10.1097/CCM.0b013
e3182 416ee 7.
67. Rech TH, Moraes RB, Crispim D, Czepielewski MA, Leitao CB. Manage‑
ment of the brain‑dead organ donor: a systematic review and meta‑
analysis. Transplantation. 2013;95:966–74. https ://doi.org/10.1097/
TP.0b013 e3182 83298 e.
68. Dhar R, Stahlschmidt E, Marklin G. A randomized trial of intravenous
thyroxine for brain‑dead organ donors with impaired cardiac function.
Prog Transplant. 2020;30:48–55. https ://doi.org/10.1177/15269 24819
89329 5.
69. Dhar R, Stahlschmidt E, Yan Y, Marklin G. A randomized trial comparing
triiodothyronine (T3) with thyroxine (T4) for hemodynamically unstable
brain‑dead organ donors. Clin Transplant. 2019;33:e13486. https ://doi.
org/10.1111/ctr.13486 .
70. Hesse UJ, Sutherland DE. Influence of serum amylase and plasma glu‑
cose levels in pancreas cadaver donors on graft function in recipients.
Diabetes. 1989;38(Suppl 1):1–3. https ://doi.org/10.2337/diab.38.1.s1.
71. Gores PF, Gillingham KJ, Dunn DL, Moudry‑Munns KC, Najarian JS,
Sutherland DE. Donor hyperglycemia as a minor risk factor and
immunologic variables as major risk factors for pancreas allograft loss
in a multivariate analysis of a single institution’s experience. Ann Surg.
1992;215:217–30. https ://doi.org/10.1097/00000 658‑19920 3000‑00005 .
72. Masson F, Thicoipe M, Gin H, de Mascarel A, Angibeau RM, Favarel‑
Garrigues JF, et al. The endocrine pancreas in brain‑dead donors. A
prospective study in 25 patients. Transplantation. 1993;56:363–7. https
://doi.org/10.1097/00007 890‑19930 8000‑00022 .
73. Odorico JS, Heisey DM, Voss BJ, Steiner DS, Knechtle SJ, D’Alessandro
AM, et al. Donor factors affecting outcome after pancreas transplanta‑
tion. Transplant Proc. 1998;30:276–7. https ://doi.org/10.1016/s0041
‑1345(97)01263 ‑3.
74. Shaffer D, Madras PN, Sahyoun AI, Simpson MA, Monaco AP. Cadaver
donor hyperglycemia does not impair long‑term pancreas allograft
survival or function. Transplant Proc. 1994;26:439–40.
75. Blasi‑Ibanez A, Hirose R, Feiner J, Freise C, Stock PG, Roberts JP, et al.
Predictors associated with terminal renal function in deceased organ
donors in the intensive care unit. Anesthesiology. 2009;110:333–41.
https ://doi.org/10.1097/ALN.0b013 e3181 94ca8 a.
76. Perez‑Protto SE, Reynolds LF, Dalton JE, Taketomi T, Irefin SA, Parker BM,
et al. Deceased donor hyperglycemia and liver graft dysfunction. Prog
Transplant. 2014;24:106–12. https ://doi.org/10.7182/pit20 14737 .
77. Sally MB, Ewing T, Crutchfield M, Patel MS, Raza S, DeLaCruz S, et al.
Determining optimal threshold for glucose control in organ donors
after neurologic determination of death: a United Network for Organ
Sharing Region 5 Donor Management Goals Workgroup prospec‑
tive analysis. J Trauma Acute Care Surg. 2014;76:62–8. https ://doi.
org/10.1097/ta.0b013 e3182 ab0d9 b.
78. Patel MS, Zatarain J, De La Cruz S, Sally MB, Ewing T, Crutchfield M, et al.
The impact of meeting donor management goals on the number of
organs transplanted per expanded criteria donor: a prospective study
from the UNOS Region 5 Donor Management Goals Workgroup. JAMA
Surg. 2014;149:969–75. https ://doi.org/10.1001/jamas urg.2014.967.
79. Khosravi MB, Firoozifar M, Ghaffaripour S, Sahmeddini MA, Eghbal MH.
Early outcomes of liver transplants in patients receiving organs from
hypernatremic donors. Exp Clin Transplant. 2013;11:537–40. https ://doi.
org/10.6002/ect.2012.0274.
80. K aseje N, McLin V, Toso C, Poncet A, Wildhaber BE. Donor hypernatremia
before procurement and early outcomes following pediatric liver
transplantation. Liver Transpl. 2015;21:1076–81. https ://doi.org/10.1002/
lt.24145 .
81. Mangus RS, Fridell JA, Vianna RM, Milgrom ML, Chestovich P, Vanden‑
boom C, et al. Severe hypernatremia in deceased liver donors does
not impact early transplant outcome. Transplantation. 2010;90:438–43.
https ://doi.org/10.1097/TP.0b013 e3181 e764c 0.
82. K aczmarek I, Tenderich G, Groetzner J, Deutsch MA, Schulz U, Beiras‑
Fernandez A, et al. The controversy of donor serum sodium levels in
heart transplantation ‑ a multicenter experience. Thorac Cardiovasc
Surg. 2006;54:313–6. https ://doi.org/10.1055/s‑2006‑92388 9.
83. Totsuka E, Dodson F, Urakami A, Moras N, Ishii T, Lee MC, et al. Influence
of high donor serum sodium levels on early postoperative graft func‑
tion in human liver transplantation: effect of correction of donor hyper‑
natremia. Liver Transpl Surg. 1999;5:421–8. https ://doi.org/10.1002/
lt.50005 0510.
84. Dawwas MF, Lewsey JD, Neuberger JM, Gimson AE. The impact of
serum sodium concentration on mortality after liver transplantation: a
cohort multicenter study. Liver Transpl. 2007;13:1115–24. https ://doi.
org/10.1002/lt.21154 .
85. Cywinski JB, Mascha E, Miller C, Eghtesad B, Nakagawa S, Vincent JP,
et al. Association between donor‑recipient serum sodium differences
Page 14 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
and orthotopic liver transplant graft function. Liver Transpl. 2008;14:59–
65. https ://doi.org/10.1002/lt.21305 .
86. Mousavi SA, Shahabi S, Mostafapour E, Purfakharan M, Fereshteh‑
nejad SM, Amini J, et al. Comparison of the serum electrolyte levels
among patients died and survived in the intensive care unit. Tanaffos.
2012;11:36–42.
87. Chen M, Sun R, Hu B. The influence of serum magnesium level
on the prognosis of critically ill patients. Zhonghua Wei Zhong
Bing Ji Jiu Yi Xue. 2015;27:213–7. https ://doi.org/10.3760/cma.j.i
ssn.2095‑4352.2015.03.011.
88. Kumar S, Honmode A, Jain S, Bhagat V. Does magnesium matter in
patients of Medical Intensive Care Unit: a study in rural Central India.
Indian J Crit Care Med. 2015;19:379–83. https ://doi.org/10.4103/0972‑
5229.16027 2.
89. Velissaris D, Karamouzos V, Pierrakos C, Aretha D, Karanikolas M.
Hypomagnesemia in critically ill sepsis patients. J Clin Med Res.
2015;7:911–8. https ://doi.org/10.14740 /jocmr 2351w .
90. Thel MC, Armstrong AL, McNulty SE, Califf RM, O’Connor CM. Ran‑
domised trial of magnesium in in‑hospital cardiac arrest. Duke Internal
Medicine Housestaff. Lancet. 1997;350:1272–6. https ://doi.org/10.1016/
s0140 ‑6736(97)05048 ‑4.
91. Smith LF, Heagerty AM, Bing RF, Barnett DB. Intravenous infusion of
magnesium sulphate after acute myocardial infarction: effects on
arrhythmias and mortality. Int J Cardiol. 1986;12:175–83. https ://doi.
org/10.1016/0167‑5273(86)90239 ‑1.
92. Alves SC, Tomasi CD, Constantino L, Giombelli V, Candal R, Bristot Mde L,
et al. Hypomagnesemia as a risk factor for the non‑recovery of the renal
function in critically ill patients with acute kidney injury. Nephrol Dial
Transplant. 2013;28:910–6. https ://doi.org/10.1093/ndt/gfs26 8.
93. Powner DJ. Factors during donor care that may affect liver transplanta‑
tion outcome. Prog Transplant. 2004;14:241–7. https ://doi.org/10.7182/
prtr.14.3.d36p8 205k2 02527 4.
94. Adam R, Reynes M, Bao YM, Astarcioglu I, Azoulay D, Chiche L, et al.
Impact of glycogen content of the donor liver in clinical liver transplan‑
tation. Transplant Proc. 1993;25:1536–7.
95. Powner DJ, Bernstein IM. Extended somatic support for pregnant
women after brain death. Crit Care Med. 2003;31:1241–9. https ://doi.
org/10.1097/01.CCM.00000 59643 .45027 .96.
96. Dominguez‑Roldan JM, Murillo‑Cabezas F, Santamaria‑Mifsut JL,
Munoz‑Sanchez A, Villen‑Nieto J, Barrera‑Chacon JM. Changes in resting
energy expenditure after development of brain death. Transplant Proc.
1995;27:2397–8.
97. Little DM, Farrell JG, Cunningham PM, Hickey DP. Donor sepsis is not
a contraindication to cadaveric organ donation. QJM. 1997;90:641–2.
https ://doi.org/10.1093/qjmed /90.10.641.
98. Zibari GB, Lipka J, Zizzi H, Abreo KD, Jacobbi L, McDonald JC. The use
of contaminated donor organs in transplantation. Clin Transplant.
2000;14:397–400. https ://doi.org/10.1034/j.1399‑0012.2000.14040 702.x.
99. Lumbreras C, Sanz F, Gonzalez A, Perez G, Ramos MJ, Aguado JM, et al.
Clinical significance of donor‑unrecognized bacteremia in the outcome
of solid‑organ transplant recipients. Clin Infect Dis. 2001;33:722–6. https
://doi.org/10.1086/32259 9.
100. Caballero F, Lopez‑Navidad A, Perea M, Cabrer C, Guirado L, Sola R.
Successful liver and kidney transplantation from cadaveric donors with
left‑sided bacterial endocarditis. Am J Transplant. 2005;5:781–7. https ://
doi.org/10.1111/j.1600‑6143.2005.00773 .x.
101. Len O, Gavalda J, Blanes M, Montejo M, San Juan R, Moreno A,
et al. Donor infection and transmission to the recipient of a solid
allograft. Am J Transplant. 2008;8:2420–5. https ://doi.org/10.111
1/j.1600‑6143.2008.02397 .x.
102. Sozen H, Fidan K, Mahli A, Singin E, Buyan N, Sindel S, et al. Successful
solid organ transplantation from septicemic cadaveric donors: case
report. Transplant Proc. 2008;40:299–301. https ://doi.org/10.1016/j.trans
proce ed.2007.11.044.
103. Lin TL, Kuo SC, Yeh CH, Chan YC, Lin YH, Li WF, et al. Donor‑transmitted
bacterial infection in deceased donor liver transplantation: experience
of Southern Taiwan Medical Center. Transplant Proc. 2018;50:2711–4.
https ://doi.org/10.1016/j.trans proce ed.2018.04.017.
104. Corman Dincer P, Tore Altun G, Birtan D, Arslantas R, Sarici Mert N,
Ozdemir I, et al. Incidence and risk factors for systemic infection
in deceased donors. Transplant Proc. 2019;51:2195–7. https ://doi.
org/10.1016/j.trans proce ed.2019.03.054.
105. Kubak BM, Gregson AL, Pegues DA, Leibowitz MR, Carlson M, Marelli
D, et al. Use of hearts transplanted from donors with severe sepsis and
infectious deaths. J Heart Lung Transplant. 2009;28:260–5. https ://doi.
org/10.1016/j.healu n.2008.11.911.
106. Outerelo C, Gouveia R, Mateus A, Cruz P, Oliveira C, Ramos A. Infected
donors in renal transplantation: expanding the donor pool. Trans‑
plant Proc. 2013;45:1054–6. https ://doi.org/10.1016/j.trans proce
ed.2013.02.014.
107. Freeman RB, Giatras I, Falagas ME, Supran S, O’Connor K, Bradley J,
et al. Outcome of transplantation of organs procured from bac‑
teremic donors. Transplantation. 1999;68:1107–11. https ://doi.
org/10.1097/00007 890‑19991 0270‑00008 .
108. Cerutti E, Stratta C, Romagnoli R, Serra R, Lepore M, Fop F, et al. Bacte‑
rial‑ and fungal‑positive cultures in organ donors: clinical impact in liver
transplantation. Liver Transpl. 2006;12:1253–9. https ://doi.org/10.1002/
lt.20811 .
109. Angelis M, Cooper JT, Freeman RB. Impact of donor infections on out‑
come of orthotopic liver transplantation. Liver Transpl. 2003;9:451–62.
https ://doi.org/10.1053/jlts.2003.50094 .
110. Ruiz I, Gavalda J, Monforte V, Len O, Roman A, Bravo C, et al. Donor‑to‑
host transmission of bacterial and fungal infections in lung trans‑
plantation. Am J Transplant. 2006;6:178–82. https ://doi.org/10.111
1/j.1600‑6143.2005.01145 .x.
111. Niemann CU, Feiner J, Swain S, Bunting S, Friedman M, Crutchfield M,
et al. Therapeutic hypothermia in deceased organ donors and kidney‑
graft function. N Engl J Med. 2015;373:405–14. https ://doi.org/10.1056/
NEJMo a1501 969.
112. Schnuelle P, Mundt HM, Druschler F, Schmitt WH, Yard BA, Kramer BK,
et al. Impact of spontaneous donor hypothermia on graft outcomes
after kidney transplantation. Am J Transplant. 2018;18:704–14. https ://
doi.org/10.1111/ajt.14541 .
113. Schnuelle P, Benck U, Kramer BK, Yard BA, Zuckermann A, Wagner F,
et al. Impact of donor core body temperature on graft survival after
heart transplantation. Transplantation. 2018;102:1891–900. https ://doi.
org/10.1097/TP.00000 00000 00233 7.
114. Huang FY, Huang BT, Wang PJ, Zuo ZL, Heng Y, Xia TL, et al. The efficacy
and safety of prehospital therapeutic hypothermia in patients with
out‑of‑hospital cardiac arrest: a systematic review and meta‑analysis.
Resuscitation. 2015;96:170–9. https ://doi.org/10.1016/j.resus citat
ion.2015.08.005.
115. Axelrod DA, Malinoski D, Patel MS, Broglio K, Lewis R, Groat T, et al. Mod‑
eling the economic benefit of targeted mild hypothermia in deceased
donor kidney transplantation. Clin Transplant. 2019;33:e13626. https ://
doi.org/10.1111/ctr.13626 .
116. de la Cruz JS, Sally MB, Zatarain JR, Crutchfield M, Ramsey K, Nielsen J,
et al. The impact of blood transfusions in deceased organ donors on
the outcomes of 1884 renal grafts from United Network for Organ Shar‑
ing Region 5. J Trauma Acute Care Surg. 2015;79:S164–70. https ://doi.
org/10.1097/TA.00000 00000 00067 0.
117. van Erp AC, van Dullemen LFA, Ploeg RJ, Leuvenink HGD. Systematic
review on the treatment of deceased organ donors. Transplant Rev
(Orlando). 2018;32:194–206. https ://doi.org/10.1016/j.trre.2018.06.001.
118. Rosendale JD, Chabalewski FL, McBride MA, Garrity ER, Rosengard
BR, Delmonico FL, et al. Increased transplanted organs from the use
of a standardized donor management protocol. Am J Transplant.
2002;2:761–8. https ://doi.org/10.1034/j.1600‑6143.2002.20810 .x.
119. Salim A, Velmahos GC, Brown C, Belzberg H, Demetriades D. Aggressive
organ donor management significantly increases the number of organs
available for transplantation. J Trauma. 2005;58:991–4. https ://doi.
org/10.1097/01.ta.00001 68708 .78049 .32.
120. Salim A, Martin M, Brown C, Rhee P, Demetriades D, Belzberg H. The
effect of a protocol of aggressive donor management: implications for
the national organ donor shortage. J Trauma. 2006;61:429–33. https ://
doi.org/10.1097/01.ta.00002 28968 .63652 .c1.
121. Malinoski DJ, Daly MC, Patel MS, Oley‑Graybill C, Foster CE, Salim A.
Achieving donor management goals before deceased donor procure‑
ment is associated with more organs transplanted per donor. J Trauma.
2011;71:990–5. https ://doi.org/10.1097/ta.0b013 e3182 2779e 5.
Page 15 of 15
Westphaletal. Ann. Intensive Care (2020) 10:169
122. Malinoski DJ, Patel MS, Daly MC, Oley‑Graybill C, Salim A, workgroup
URD. The impact of meeting donor management goals on the number
of organs transplanted per donor: results from the United Network for
Organ Sharing Region 5 prospective donor management goals study.
Crit Care Med. 2012;40:2773–80. https ://doi.org/10.1097/ccm.0b013
e3182 5b252 a.
123. Marshall GR, Mangus RS, Powelson JA, Fridell JA, Kubal CA, Tector AJ.
Donor management parameters and organ yield: single center results. J
Surg Res. 2014;191:208–13. https ://doi.org/10.1016/j.jss.2014.02.054.
124. Patel MS, De La Cruz S, Sally MB, Groat T, Malinoski DJ. Active donor
management during the hospital phase of care is associated with more
organs transplanted per donor. J Am Coll Surg. 2017;225:525–31. https
://doi.org/10.1016/j.jamco llsur g.2017.06.014.
125. Malinoski DJ, Patel MS, Ahmed O, Daly MC, Mooney S, Graybill CO, et al.
The impact of meeting donor management goals on the develop‑
ment of delayed graft function in kidney transplant recipients. Am J
Transplant. 2013;13:993–1000. https ://doi.org/10.1111/ajt.12090 .
126. Westphal GA, Zaclikevis VR, Vieira KD, Cordeiro Rde B, Horner MB,
Oliveira TP, et al. A managed protocol for treatment of deceased
potential donors reduces the incidence of cardiac arrest before organ
explant. Rev Bras Ter Intensiva. 2012;24:334–40. https ://doi.org/10.1590/
s0103 ‑507x2 01200 04000 07.
127. Haynes AB, Weiser TG, Berry WR, Lipsitz SR, Breizat AH, Dellinger EP, et al.
A surgical safety checklist to reduce morbidity and mortality in a global
population. N Engl J Med. 2009;360:491–9. https ://doi.org/10.1056/
NEJMs a0810 119.
128. Pronovost P, Needham D, Berenholtz S, Sinopoli D, Chu H, Cosgrove
S, et al. An intervention to decrease catheter‑related bloodstream
infections in the ICU. N Engl J Med. 2006;355:2725–32. https ://doi.
org/10.1056/NEJMo a0611 15.
129. Weiss CH, Moazed F, McEvoy CA, Singer BD, Szleifer I, Amaral LA, et al.
Prompting physicians to address a daily checklist and process of care
and clinical outcomes: a single‑site study. Am J Respir Crit Care Med.
2011;184:680–6. https ://doi.org/10.1164/rccm.20110 1‑0037O C.
130. Writing Group for the C‑ICUI, the Brazilian Research in Intensive Care,
Cavalcanti AB, Bozza FA, Machado FR, Salluh JI, et al. Effect of a quality
improvement intervention with daily round checklists, goal setting,
and clinician prompting on mortality of critically ill patients: a rand‑
omized clinical trial. JAMA. 2016;315:1480–90. https ://doi.org/10.1001/
jama.2016.3463.
131. Ball IM, Hornby L, Rochwerg B, Weiss MJ, Gillrie C, Chasse M, et al. Man‑
agement of the neurologically deceased organ donor: a Canadian clini‑
cal practice guideline. CMAJ. 2020;192:E361–9. https ://doi.org/10.1503/
cmaj.19063 1.
132. Meyfroidt G, Gunst J, Martin‑Loeches I, Smith M, Robba C, Taccone FS,
et al. Management of the brain‑dead donor in the ICU: general and
specific therapy to improve transplantable organ quality. Intensive Care
Med. 2019;45:343–53. https ://doi.org/10.1007/s0013 4‑019‑05551 ‑y.
133. National Academies of Sciences, Engineering, and Medicine. Opportu‑
nities for organ donor intervention research: Saving lives by improving
the quality and quantity of organs for transplantation. Washington: The
National Academies Press; 2017.
134. Helms AK, Torbey MT, Hacein‑Bey L, Chyba C, Varelas PN. Standardized
protocols increase organ and tissue donation rates in the neurocriti‑
cal care unit. Neurology. 2004;63:1955–7. https ://doi.org/10.1212/01.
wnl.00001 44197 .06562 .24.
135. Franklin GA, Santos AP, Smith JW, Galbraith S, Harbrecht BG, Garrison
RN. Optimization of donor management goals yields increased organ
use. Am Surg. 2010;76:587–94.
136. Westphal GA, Robinson CC, Biasi A, Machado FR, Rosa RG, Teixeira C,
et al. DONORS (Donation Network to Optimise Organ Recovery Study):
Study protocol to evaluate the implementation of an evidence‑based
checklist for brain‑dead potential organ donor management in inten‑
sive care units, a cluster randomised trial. BMJ Open. 2019;9:e028570.
https ://doi.org/10.1136/bmjop en‑2018‑02857 0.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub‑
lished maps and institutional affiliations.
... Die Organdysfunktion beim Spender ist einer der Hauptgründe, warum eine Transplantation trotz Zustimmung nicht realisiert werden kann [8,43]. Den Intensivmedizinern stehen für die organprotektiven intensivmedizinischen Maßnahmen internationale Leitlinien [2,27,46] sowie nationale Empfehlungen zur Verfügung [13,24,29]. ...
... Die zugrunde liegenden pathophysiologischen Veränderungen sind immer noch nicht komplett verstanden. Zur detaillierten Beschreibung der Vorgänge sei auf die entsprechende Literatur verwiesen [24,27,32,46]. Eine Zusammenfassung der Empfehlungen zu organprotektiven Maßnahmen zeigt . ...
... Eine Zusammenfassung der Empfehlungen zu organprotektiven Maßnahmen zeigt . Tab. 1. Diese Empfehlungen sind abgeleitet aus den vorhandenen internationalen Leitlinien [2,27,32,34,46] und nationalen Empfehlungen [13,24,29], grundsätzlich ist jedoch die Evidenz für alle Empfehlungen aufgrund fehlender randomisierter kontrollierter Studien (RCT) gering [32,35]. ...
Article
Full-text available
Zusammenfassung Hintergrund Die Anzahl postmortal gespendeter Organe ist in Deutschland weit geringer als der Bedarf. Dies unterstreicht die Wichtigkeit einer optimalen Versorgung während des gesamten Prozesses der Organspende. Fragestellung Es existieren internationale Leitlinien und nationale Empfehlungen zu intensivmedizinischen organprotektiven Maßnahmen beim Organspender. Für das anästhesiologische Management fehlen evidenzbasierte Empfehlungen. Ziel dieser Übersichtsarbeit ist es, anhand der vorhandenen Evidenz die pathophysiologischen Veränderungen des irreversiblen Hirnfunktionsausfalls zu rekapitulieren und sich kritisch mit den empfohlenen Behandlungsstrategien und therapeutischen Zielgrößen auseinanderzusetzen. Auch auf ethische Aspekte der Betreuung des postmortalen Organspenders wird eingegangen. Methode Diese Übersichtsarbeit basiert auf einer selektiven Literaturrecherche in PubMed (Suchwörter: „brain dead donor“, „organ procurement“, „organ protective therapy“, „donor preconditioning“, „perioperative donor management“, „ethical considerations of brain dead donor“). Internationale Leitlinien und nationale Empfehlungen wurden besonders berücksichtigt. Ergebnisse Insgesamt ist die Evidenz für optimale intensivmedizinische und perioperative organprotektive Maßnahmen beim postmortalen Organspender sehr gering. Nationale und internationale Empfehlungen zu Zielwerten und medikamentösen Behandlungsstrategien unterscheiden sich teilweise erheblich: kontrollierte randomisierte Studien fehlen. Der Stellenwert einer Narkose zur Explantation bleibt sowohl unter pathophysiologischen Gesichtspunkten als auch aus ethischer Sicht ungeklärt. Schlussfolgerungen Die Kenntnisse über die pathophysiologischen Prozesse im Rahmen des irreversiblen Hirnfunktionsausfalls und die organprotektiven Maßnahmen sind ebenso Grundvoraussetzung wie die ethische Auseinandersetzung mit dem Thema postmortale Organspende. Nur dann kann das Behandlungsteam in dieser herausfordernden Situation sowohl dem Organempfänger als auch dem Organspender und seinen Angehörigen gerecht werden.
... O tempo entre a confirmação da ME e a PCR corresponde ao tempo em que o PD ficou aos cuidados da equipe multiprofissional da Unidade de Terapia Intensiva (UTI), ou seja, é realizada a manutenção do PD, cujo objetivo é assegurar as funções orgânicas estáveis e os órgãos aptos ao transplante 22,23 . Nesse estudo a inexistência de registros do tempo entre a confirmação da ME e a PCR ficou próximo a 60% da amostra das fichas analisadas. ...
... Quanto ao aspecto clínico de manutenção do PD, afirma-se que o tempo seja o fator determinante para o sucesso e interferência direta na qualidade dos enxertos e consequentemente a efetivação do transplante de órgãos e tecidos, bem como na avaliação desse processo institucionalmente 21,23 . Isto posto, pode-se estar associado à dificuldade de os profissionais médicos determinarem o horário legal do óbito, resultado encontrado em um estudo que objetivou avaliar o conhecimento de 90 médicos atuantes em UTI sobre ME 24 . ...
Article
Full-text available
Objective to analyze the epidemiological characteristics and causes of non-completion of the organ and tissue donation process from potential brain-dead donors. Method cross-sectional study, with secondary data from 2019, obtained from brain death notification forms of patients aged 18 years and over, analyzed descriptively and inferentially, after approval by the ethics committee. Results 231 brain deaths were reported, with a higher incidence of males, average age of 48 years and notifications from public hospitals. In Campo Grande, Mato grosso do Sul, Brazil, there was a greater number of notifications and a shorter time between notification and the first clinical examination (RR: 4.01; CI 2.17-7.41; p<0.001). Among cases of non-donation, 75.8% occurred due to medical contraindication and family refusal. Conclusion there was a predominance of young adults, non-organ donors, whose family's refusal was due to the desire to keep the body intact.
... [2] Thus, subsequent studies have established a hemoglobin concentration of 7.0 g/dL as the threshold for red blood cell (RBC) transfusion. [3,4] Guidelines for the management of BDODs consist of ventilator care, hemodynamic support, hormonal resuscitation, and the correction of metabolic derangement; however, most goals were derived from studies on patients experiencing shock and not BDODs. A recommended level of hemoglobin concentration has been inferred from shock resuscitation studies without verification on the BDOD population, despite a substantial effect on organ function. ...
... [10,11] The transfusion threshold in BDODs has been gradually shifted toward a hemoglobin concentration of 7 g/dL since the TRICC trial demonstrated the superiority of restrictive transfusion strategy. [3,4,12] However, studies on transfusion threshold, including the TRICC trial, were conducted in critically ill patients and not BDODs, therefore, guidelines for the transfusion threshold for the BDOD population has not been established. Although restrictive transfusion strategy has been widely accepted, RBC transfusion was required in 63.2% and 15.8% of BDODs with trough hemoglobin concentrations of > 8 g/ dL and > 10 g/dL, respectively, in a study that demonstrated the actual practice of the transfusion. ...
Article
Full-text available
The disparity between the demand and supply of organs has necessitated an expansion of the criteria for organ donation. Consequently, numerous guidelines have been proposed for managing brain-dead organ donors (BDODs) to improve their organ function and the organ procurement rate. Therefore, we aimed to evaluate the previously recommended threshold for red blood cell transfusion in BDODs. Medical records of BDODs were retrospectively reviewed from January 2012 to December 2021. We enrolled BDODs who stayed for more than 24 hours at an hospital organ procurement organization. We analyzed their organ function and the rate of organ procurement according to the hemoglobin concentration. A total of 111 BDODs were enrolled and divided into the following 2 groups: hemoglobin (Hb) ≥ 10 g/dL (45.0 %) and Hb < 10 g/dL (55.0 %). There were no significant differences between the groups in the total bilirubin, creatinine, arterial blood lactate, and the rate of organ procurement. A correlation analysis did not reveal any association between the hemoglobin concentration and organ function of the BDODs. Hemoglobin concentration of 10 g/dL cannot be considered a threshold for red blood cell transfusion. Furthermore, organ function is not correlated with a hemoglobin concentration > 7 g/dL. Restrictive transfusion strategy is appropriate for BDOD management.
... The management of brain-dead donors was discrete and conducted by the critical care team in accordance with local standards [19][20][21] . They were unaware of study Plasma IL-6, IL-1β, IL-10, IFN-, TNF, and BCL-2 Results are expressed as pg/mL. ...
Article
Background Brain death triggers an inflammatory cascade that damages organs before procurement, adversely affecting the quality of grafts. This randomized clinical trial aimed to compare the efficacy of liraglutide compared to placebo in attenuating brain death-induced inflammation, endoplasmic reticulum stress, and oxidative stress. Methods We conducted a double-blinded, placebo-controlled, randomized clinical trial with brain-dead donors. Fifty brain-dead donors were randomized to receive subcutaneous liraglutide or placebo. The primary outcome was the reduction in interleukin-6 (IL-6) plasma levels. Secondary outcomes were changes in other plasma pro-inflammatory (IL-1β, IFN-γ, TNF) and anti-inflammatory cytokines (IL-10), expression of antiapoptotic ( BCL2 ), endoplasmic reticulum stress markers ( DDIT3/CHOP , HSPA5/BIP ), and antioxidant ( SOD2 , UCP2 ) genes, and expression TNF, DDIT3, and SOD2 proteins in liver biopsies. Results The liraglutide group showed lower cytokine levels compared to the placebo group during follow-up: Δ IL-6 (−28 [−182–135] vs. 32 [−10.6–70.7] pg/mL; p =0.041) and Δ IL-10 (−0.01 [−2.2–1.5] vs. 1.9 [−0.2–6.1] pg/mL; p =0.042), respectively. The administration of liraglutide did not significantly alter the expression of inflammatory, anti-apoptotic, endoplasmic reticulum stress, or antioxidant genes in the liver tissue. Similar to gene expression, expressions of proteins in liver were not affected by the administration of liraglutide. Treatment with liraglutide did not increase organ recovery rate [OR=1.2 (95% CI 0.2–8.6), p =0.82]. Conclusions Liraglutide administration reduced IL-6 and prevented the increase of IL-10 plasma levels in brain-dead donors, without affecting the expression of genes and proteins related to inflammation, apoptosis, endoplasmic reticulum stress, or oxidative stress.
... Potansiyel donörde gerçekleşen enflamatuvar süreç ve %75 oranında görülen adrenal yetmezlik sebebiyle hidrokortizon 300 mg iv, sonra her sekiz saatte bir 100 mg önerilmektedir (12,26). CORTICOME çalışmasında hidrokortizon verilen grupta noradrenalin dozunun daha düşük olduğu ve hemodinamik stabilitenin daha çabuk sağlandığı saptanmıştır (27). ...
... In usual clinical practice, brain-dead patients are validated as potential donors based on the medical history, vital data over time, and laboratory tests that can demonstrate target organ damage [2,34,35]. In the present study, we assessed three different potential therapeutic donor care strategies (groups B, C, and D), commonly used in clinical practice, and verified the effect on parameters routinely used for the acceptance of an organ, which reflects the external validity. ...
Article
Full-text available
Introduction: Most transplanted organs are obtained from brain-dead donors. Inflammation results in a higher rate of rejection. Objectives: The objective of this animal model of brain death (BD) was to evaluate the effect of the progressive institution of volume expansion, norepinephrine, and combined hormone therapy on clinical, laboratory, and histological aspects. Methods: Twenty rabbits were divided: A (control), B (induction of BD + infusion of crystalloid), C (BD + infusion of crystalloid and noradrenaline (NA)), and D (BD + infusion of crystalloid + vasopressin + levothyroxine + methylprednisolone + NA). The animals were monitored for four hours with consecutives analysis of vital signs and blood samples. The organs were evaluated by a pathologist. Results: In Group D, we observed fewer number and lesser volume of infusions (p = 0.032/0.014) when compared with groups B and C. Mean arterial pressure levels were higher in group D when compared with group B (p = 0.008). Group D had better glycemic control when compared with group C (p = 0.016). Sodium values were elevated in group B in relation to groups C and D (p = 0.021). In Group D, the organ perfusion was better. Conclusion: The optimized strategy of management of BD animals is associated with better hemodynamic, glycemic, and natremia control, besides reducing early signs of ischemia.
... (13) In view of this, a task force of societies and associations linked to organ transplantation in Brazil sought to develop and disseminate protocols for managing the potential donor in the ICU. (14) Although crucial, optimization of care of the potential donor cannot change the patient's biological characteristics, such as age, weight, and comorbidities, nor reverse previous aggressions to the organs. ...
Article
Full-text available
Objective: To understand the professionals´ perception of the use of deceased donor liver for transplantation, the reasons to decline them, and propose strategies to increase their use safely. Methods: This is a cross-sectional, descriptive qualitative-quantitative study. Professionals working with liver transplantation answered a self-administered, structured, and electronic questionnaire comprising 17 questions distributed into four sessions (demographic factors, perception of use of organs, reasons for disposal, and measures to favor their usage). Results: A total of 42 professionals participated in the study. The rate of use of organs was considered low by 71.43% (n=30) of respondents or very low by 19.05% (n=8). Everyone agreed that it was possible to increase it. Thirty-one (73.81%) participants believed the expansion of the population of extended criteria donors affected this index negatively. Donor-related conditions were the most frequent category of reasons for refusing a liver for transplantation, being the findings during organ retrieval the most frequent reason in clinical practice. Enhanced training of intensive care teams in the treatment of donors was the primary measure selected to favor the use of the organs, followed by investment in new technologies to optimize its preservation/evaluate its function before transplantation. Conclusion: Implementation of strategies to increase the rate of acceptance of livers is expected. Improvements in donor intensive care and implementation of new preservation technologies should favor the use of the organs.
Article
Full-text available
Importance The effectiveness of goal-directed care to reduce loss of brain-dead potential donors to cardiac arrest is unclear. Objective To evaluate the effectiveness of an evidence-based, goal-directed checklist in the clinical management of brain-dead potential donors in the intensive care unit (ICU). Design, Setting, and Participants The Donation Network to Optimize Organ Recovery Study (DONORS) was an open-label, parallel-group cluster randomized clinical trial in Brazil. Enrollment and follow-up were conducted from June 20, 2017, to November 30, 2019. Hospital ICUs that reported 10 or more brain deaths in the previous 2 years were included. Consecutive brain-dead potential donors in the ICU aged 14 to 90 years with a condition consistent with brain death after the first clinical examination were enrolled. Participants were randomized to either the intervention group or the control group. The intention-to-treat data analysis was conducted from June 15 to August 30, 2020. Interventions Hospital staff in the intervention group were instructed to administer to brain-dead potential donors in the intervention group an evidence-based checklist with 13 clinical goals and 14 corresponding actions to guide care, every 6 hours, from study enrollment to organ retrieval. The control group provided or received usual care. Main Outcomes and Measures The primary outcome was loss of brain-dead potential donors to cardiac arrest at the individual level. A prespecified sensitivity analysis assessed the effect of adherence to the checklist in the intervention group. Results Among the 1771 brain-dead potential donors screened in 63 hospitals, 1535 were included. These patients included 673 males (59.2%) and had a median (IQR) age of 51 (36.3-62.0) years. The main cause of brain injury was stroke (877 [57.1%]), followed by trauma (485 [31.6%]). Of the 63 hospitals, 31 (49.2%) were assigned to the intervention group (743 [48.4%] brain-dead potential donors) and 32 (50.8%) to the control group (792 [51.6%] brain-dead potential donors). Seventy potential donors (9.4%) at intervention hospitals and 117 (14.8%) at control hospitals met the primary outcome (risk ratio [RR], 0.70; 95% CI, 0.46-1.08; P = .11). The primary outcome rate was lower in those with adherence higher than 79.0% than in the control group (5.3% vs 14.8%; RR, 0.41; 95% CI, 0.22-0.78; P = .006). Conclusions and Relevance This cluster randomized clinical trial was inconclusive in determining whether the overall use of an evidence-based, goal-directed checklist reduced brain-dead potential donor loss to cardiac arrest. The findings suggest that use of such a checklist has limited effectiveness without adherence to the actions recommended in this checklist. Trial Registration ClinicalTrials.gov Identifier: NCT03179020
Article
Full-text available
Objective To collate and summarise the literature on the quality improvement tools that have been developed for deceased organ donation processes after circulatory determination of death and neurological determination of death. Design Scoping review using the Joanna Briggs Institute framework. Data sources We searched for published (MEDLINE, Embase, PsycINFO, CINAHL, Web of Science) and unpublished literature (organ donation organisation websites worldwide). The search was initially conducted on 17 July 2021 and updated on 1 June 2022. Included articles discussed the creation and/or use of quality improvement tools to manage deceased organ donation processes. Two independent reviewers screened the references, extracted and analysed the data. Results 40 references were included in this review, and most records were written in English (n=38), originated in Canada (n=21), published between 2016 and 2022 (n=22), and were specific for donation after neurological determination of death (n=20). The tools identified included checklists, algorithms, flow charts, charts, pathways, decision tree maps and mobile apps. These tools were applied in the following phases of the organ donation process: (1) potential donor identification, (2) donor referral, (3) donor assessment and risk, (4) donor management, (5) withdrawal of life-sustaining measures, (6) death determination, (7) organ retrieval and (8) overall organ donation process. Conclusions We conducted a thorough investigation of the available quality improvement tools for deceased organ donation processes. The existing evidence lacks details in the report of methods used for development, testing and impact of these tools, and we could not locate tools specific for some phases of the organ donation process. Lastly, by mapping existing tools, we aim to facilitate both clinician choices among available tools, as well as research work building on existing knowledge.
Article
Full-text available
Introduction There is an increasing demand for multi-organ donors for organ transplantation programmes. This study protocol describes the Donation Network to Optimise Organ Recovery Study, a planned cluster randomised controlled trial that aims to evaluate the effectiveness of the implementation of an evidence-based, goal-directed checklist for brain-dead potential organ donor management in intensive care units (ICUs) in reducing the loss of potential donors due to cardiac arrest. Methods and analysis The study will include ICUs of at least 60 Brazilian sites with an average of ≥10 annual notifications of valid potential organ donors. Hospitals will be randomly assigned (with a 1:1 allocation ratio) to the intervention group, which will involve the implementation of an evidence-based, goal-directed checklist for potential organ donor maintenance, or the control group, which will maintain the usual care practices of the ICU. Team members from all participating ICUs will receive training on how to conduct family interviews for organ donation. The primary outcome will be loss of potential donors due to cardiac arrest. Secondary outcomes will include the number of actual organ donors and the number of organs recovered per actual donor. Ethics and dissemination The institutional review board (IRB) of the coordinating centre and of each participating site individually approved the study. We requested a waiver of informed consent for the IRB of each site. Study results will be disseminated to the general medical community through publications in peer-reviewed medical journals. Trial registration number NCT03179020 ; Pre-results.
Article
Full-text available
Purpose To provide a practical overview of the management of the potential organ donor in the intensive care unit. Methods Seven areas of donor management were considered for this review: hemodynamic management; fluids and electrolytes; respiratory management; endocrine management; temperature management; anaemia and coagulation; infection management. For each subchapter, a narrative review was conducted. Results and conclusions Most elements in the current recommendations and guidelines are based on pathophysiological reasoning, epidemiological observations, or extrapolations from general ICU management strategies, and not on evidence from randomized controlled trials. The cardiorespiratory management of brain-dead donors is very similar to the management of critically ill patients, and the same applies to the management of anaemia and coagulation. Central diabetes insipidus is of particular concern, and should be diagnosed based on clinical criteria. Depending on the degree of vasopressor dependency, it can be treated with intermittent desmopressin or continuous vasopressin, intravenously. Temperature management of the donor is an area of uncertainty, but it appears reasonable to strive for a core temperature of > 35 °C. The indications and controversies regarding endocrine therapies, in particular thyroid hormone replacement therapy, and corticosteroid therapy, are discussed. The potential donor should be assessed clinically for infections, and screening tests for specific infections are an essential part of donor management. Although the rate of infection transmission from donor to receptor is low, certain infections are still a formal contraindication to organ donation. However, new antiviral drugs and strategies now allow organ donation from certain infected donors to be done safely.
Article
Full-text available
Background: Currently, there is no consensus on which treatments should be a part of standard deceased-donor management to improve graft quality and transplantation outcomes. The objective of this systematic review was to evaluate the effects of treatments of the deceased, solid-organ donor on graft function and survival after transplantation. Methods: Pubmed, Embase, Cochrane, and Clinicaltrials.gov were systematically searched for randomized controlled trials that compared deceased-donor treatment versus placebo or no treatment. Results: A total of 33 studies were selected for this systematic review. Eleven studies were included for meta-analyses on three different treatment strategies. The meta-analysis on methylprednisolone treatment in liver donors (two studies, 183 participants) showed no effect of the treatment on rates of acute rejection. The meta-analysis on antidiuretic hormone treatment in kidney donors (two studies, 222 participants) indicates no benefit in the prevention of delayed graft function. The remaining meta-analyses (seven studies, 334 participants) compared the effects of 10 min of ischaemic preconditioning on outcomes after liver transplantation and showed that ischaemic preconditioning improved short-term liver function, but not long-term transplant outcomes. Conclusions: There is currently insufficient evidence to conclude that any particular drug treatment or any intervention in the deceased donor improves long-term graft or patient survival after transplantation.
Article
Rationale Brain death (BD) precipitates cardiac dysfunction impairing the ability to transplant hearts from eligible organ donors. Retrospective studies have suggested that thyroid hormone may enhance myocardial recovery and increase hearts transplanted. We performed a randomized trial evaluating whether intravenous thyroxine (T4) improves cardiac function in BD donors with impaired ejection fraction (EF). Methods All heart-eligible donors managed at a single-organ procurement organization (OPO) underwent protocolized fluid resuscitation. Those weaned off vasopressors underwent transthoracic echocardiography (TTE) within 12 hours of BD and, if EF was below 60%, were randomized to T4 infusion or no T4 for 8 hours, after which TTE was repeated. Results Of 77 heart-eligible donors, 36 were weaned off vasopressors. Ejection fraction was depressed in 30, of whom 28 were randomized to T4 (n = 17) vs control (n = 11). Baseline EF was comparable (45%, interquartile range [IQR] 42.5-47.5 vs 40%, 40-50, P = .32). Ejection fraction did not improve more with T4 (10%, IQR 5-15 vs 5%, 0-12.5, P = .24), although there was a trend to more hearts transplanted (59% vs 27%, P = .14). This difference appeared to be accounted for by more donors with a history of drug use in the T4 group, who exhibited greater improvements in EF (15% vs 0% without drug use, P = .01) and more often had hearts transplanted (12 of 19 vs 1 of 9, P = .01). Conclusions In this small randomized study of BD donors with impaired cardiac function, T4 infusion did not result in greater cardiac recovery. A larger randomized trial comparing T4 to placebo appears warranted but would require collaboration across multiple OPOs.
Article
Background: Organ donation shortage is the primary barrier to all organ transplantations.Infectious disease transmission through transplantation is considered controversial for organ retrieval. Donors with bacteremia and sepsis are considered controversial for organ retrieval due to potential transmission of an infectious agent to the recipient. Methods: We retrospectively reviewed the results of bacterial culture of the donor's blood from peripheral venous or central venous catheter, urine, and bronchial aspiration from the organ donation registries of 102 potential donors from the Ministry of Health and Tissue Transplant Coordination Center of Istanbul Region in 2015. Results: Of the 102 deceased donors included in the analysis, 24 (23.5%) had infection. The most common sites of infection were the bloodstream (41.6%) and the respiratory system (37.5%). The most common isolated pathogens of the bacterial cultures were Gram-positive bacteria (21), Gram-negative microorganisms (14), and Candida (1). The significant risk factor for infection was duration of stay at the intensive care unit (median: 5 day; 25-75%: 3-5 day) (odds ratio, 2.94; 95% confidence interval, 1.06-8.12; P < .05). The presence of infection in the donor accounted for a significant part of the reasons why the organs were not accepted for transplantation (kidneys 9%, liver 4%, heart 6%). Conclusions: The study showed that deceased donors with prolonged stays in the intensive care unit have an increased risk for developing nosocomial infections; so there is a need for establishing and enforcing the prevention and control of infection in possible donors.
Article
Delayed graft function (DGF) in kidney transplant significantly increases inpatient and outpatient cost. Targeted, mild hypothermia in organ donors after neurologic determination of death significantly reduced the rate of DGF in a recent randomized controlled clinical trial. To assess the potential economic benefit of national implementation of donor hypothermia, rates of reduction DGF were combined with estimates of the impact of DGF on hospital cost and total health expenditure for standard and extended criteria donor organs (SCD and ECD). DGF increases the cost of the transplant episode by $9,487 for ECD transplant and $10,342 for SCD transplant. Medicare recipients with DGF incur an additional $18,513 spending for ECD and $14,948 in SCD transplants over the first year. An absolute reduction in DGF rate after kidney transplantation consistent with trial results (ECD 25%, SCD 7%) has the potential to lower annual hospital cost for kidney transplant by $13,178,746 and annual Medicare spending by $20,970,706 compared to standard donor management practice using static cold storage. Targeted mild hypothermia improves care of renal transplant patients by safely reducing DGF rates in both ECD and SCD transplant. Broader application of this safe, effective, and low‐cost intervention could reduce health care expenditures for providers and insurers. This article is protected by copyright. All rights reserved.
Article
Rationale Brain‐dead (BD) organ donors frequently exhibit hemodynamic instability and/or reversible cardiac dysfunction. Retrospective studies have suggested that thyroid hormone may stabilize hemodynamics and enhance myocardial recovery. Intravenous levothyroxine (T4) is most frequently utilized but studies have suggested that triiodothyronine (T3) may be superior. We performed a randomized comparative‐effectiveness trial to address this uncertainty in donor management. Methods All heart‐eligible donors managed at a single OPO underwent standardized fluid resuscitation. If not weaned off vasopressors, donors underwent echocardiography (within 12 hours of BD) and were randomized to T3 or T4 infusion for eight hours. Results 37 BD donors were randomized (16 T3 vs. 21 T4). Baseline ejection fraction (EF) was comparable (median 38% vs. 45%, p=0.87) as was vasopressor dosage (6 vs. 12 μg/min of norepinephrine, NE, p=0.12). Reduction in NE dose and proportion weaned off vasopressors was similar and LVEF improved in both groups (repeat EF: 50% vs. 52.5%, p=0.38) with almost half attaining EF ≥ 55%. Although more hearts were transplanted in the T3 group (10/16 vs. 6/21, p=0.04), this difference did not persist after adjusting for baseline imbalances in age and PF ratio. Conclusions Infusion of T3 does not appear to confer significant hemodynamic or cardiac benefits over T4 for hemodynamic unstable BD organ donors. This article is protected by copyright. All rights reserved.
Article
Treatment of the brain-dead donor (DBD) with low-dose dopamine improves the outcomes after kidney and heart transplantation. This study investigates the course of liver allografts from multi-organ donors enrolled in the randomized dopamine trial between 2004 and 2007 (clinicalTrials. gov Identifier: NCT00115115). Twohundred sixty-four hemodynamically stable DBDs were randomly assigned to receive low-dose dopamine. Dopamine was infused at 4 μg/kg/min for a median duration of 6.0 hours (IQR 4.4-7.5 hours). We assessed the outcomes of 212 liver transplants performed at 32 European centers. Donors and recipients of both groups were very similar in baseline characteristics. Pretransplant labMELD-score was not different in recipients of a dopamine-treated vs. untreated graft (18 ± 8 vs. 20 ± 8; P=0.12). Mean cold ischemic time was 10.6 ± 2.9 vs. 10.1 ± 2.8 hours; P=0.24. No differences occurred in biopsy-proven rejection episodes (14.4% vs. 15.7%; P=0.85), requirement of hemofiltration (27.9% vs. 31.5%; P=0.65), the need for early re-transplantation (5.8% vs. 6.5%; P>0.99), the incidence of primary non-function (7.7% vs. 8.3%; P>0.99), and in-hospital mortality (15.4% vs. 14.8%; P>0.99). Graft survival was 71.2% vs. 73.2% and 59.6% vs. 62.0% at one and three years; Log rank P=0.71. Patient survival was 76.0% vs. 78.7% and 65.4% vs. 69.4% at one and three years; Log rank P=0.50. Conclusion: Donor pretreatment with dopamine has no short or long term effects on outcome after liver transplantation. Therefore, low-dose dopamine pretreatment can safely be implemented as the standard of care in hemodynamically stable DBDs. This article is protected by copyright. All rights reserved.
Article
Background: A previous donor intervention trial found that induction of mild therapeutic hypothermia in the brain-dead donor reduced the dialysis requirement after kidney transplantation. Consequences on the performance of cardiac allografts after transplantation were not explored to date. Methods: Cohort study investigating 3-year heart allograft survival according to spontaneous core body temperature (CBT) assessed on the day of organ procurement. The study is nested in the database of the randomized trial of donor pretreatment with low-dose dopamine (ClinicalTrials.gov identifier: NCT000115115). Results: 99 heart transplant recipients who had received a cardiac allograft from a multiorgan donor enrolled in the dopamine trial were grouped by tertiles of the donor's CBT assessed by a mere temperature reading 4-20 hours before procurement (lowest, 32.0-36.2°C; middle, 36.3-36.8°C; highest, 36.9-38.8°C). Baseline characteristics considering demographics of donors and recipients, concomitant donor treatments, donor hemodynamic and respiratory parameters as well as underlying cardiac diseases in recipients, pretransplant hemodynamic assessments, including pretransplant inotropic / mechanical support, urgency, and waiting-time were similar. A lower CBT was associated with inferior heart allograft survival, hazard ratio (HR) 0.53, 95% confidence interval (CI) 0.31-0.93, per tertile; p=0.02, and HR 0.68, 95% CI 0.50-0.93, per degree Celsius; p=0.02, when CBT was included as continuous explanatory variable in the Cox regression analysis. Conclusions: A lower CBT in the brain-dead donor before procurement may associate with an unfavorable clinical course after heart transplantation. More research is required, before therapeutic hypothermia can routinely be used in multiorgan donors when a cardiac transplantation is intended.