You are on page 1of 14

Position Statement

A Roadmap for Innovation to Advance Transplant Access


and Outcomes: A Position Statement From the National
Kidney Foundation
Krista L. Lentine, Stephen Pastan, Sumit Mohan, Peter P. Reese, Alan Leichtman, Francis L. Delmonico,
Gabriel M. Danovitch, Christian P. Larsen, Lyndsay Harshman, Alexander Wiseman, Holly J. Kramer,
Joseph Vassalotti, Jessica Joseph, Kevin Longino, Matthew Cooper,* and David A. Axelrod,* on behalf of the
National Kidney Foundation

Over the past 65 years, kidney transplantation has evolved into the optimal treatment for patients with kidney Complete author and article
failure, dramatically reducing suffering through improved survival and quality of life. However, access to trans- information provided before
references.
plant is still limited by organ supply, opportunities for transplant are inequitably distributed, and lifelong transplant
survival remains elusive. To address these persistent needs, the National Kidney Foundation convened an expert Correspondence to
panel to define an agenda for future research. The key priorities identified by the panel center on the needs to K.L. Lentine (krista.lentine@
health.slu.edu)
develop and evaluate strategies to expand living donation, improve waitlist management and transplant readi-
ness, maximize use of available deceased donor organs, and extend allograft longevity. Strategies targeting the *M.C. and D.A.A. contributed
critical goal of decreasing organ discard that warrant research investment include educating patients and cli- equally to this work.
nicians about potential benefits of accepting nonstandard organs, use of novel organ assessment technologies Am J Kidney Dis.
and real-time decision support, and approaches to preserve and resuscitate allografts before implantation. The 78(3):319-332. Published
development of personalized strategies to reduce the burden of lifelong immunosuppression and support “one online July 27, 2021.
transplant for life” was also identified as a vital priority. The panel noted the specific goal of improving transplant doi: 10.1053/
access and graft survival for children with kidney failure. This ambitious agenda will focus research investment to j.ajkd.2021.05.007
promote greater equity and efficiency in access to transplantation, and help sustain long-term benefits of the gift © 2021 by the National
of life for more patients in need. Kidney Foundation, Inc.

As this article reflects the official position of the National


potentially transplantable organs continue to go unused.
Kidney Foundation (NKF) and because it was reviewed and Decisions not to use recovered organs are generally
approved by NKF, it was not peer reviewed by AJKD. This attributed to kidney quality, but are, in fact, multifactorial,
article was prepared by a work group comprising the authors reflecting opinions about transplant benefit, cost, program
and chaired by Dr Krista Lentine. It was reviewed and regulatory risk, and logistical complexity, not all of which
approved by the NKF Scientific Advisory Board and the NKF are supported by evidence-based assessments.
Executive Committee. The population in need of transplant is increasingly
complex and diverse, including a higher prevalence of
older patients, as well as individuals with comorbidities
(eg, obesity, diabetes mellitus, cardiac disease) and of non-
Introduction White race or ethnicity (Fig 1).2 Furthermore, rates of
Kidney transplantation—and especially living-donor kid- LDKT in adults and children have generally stagnated while
ney transplantation (LDKT)—offers patients with kidney racial disparities in LDKT access persist or worsen.5 The
failure the best chance for long-term survival and need to increase access to kidney transplantation received
improved quality of life, at the lowest costs to the health unprecedented support from the federal government in the
care system.1,2 There have been many positive trends in July 2019 Advancing American Kidney Health executive
kidney transplantation in recent years, including a pro- order, which articulated goals for increasing the initiation
gressive rise in rates of deceased donor kidney trans- of kidney replacement therapy through transplant and
plantation and improvements in short- and longer-term substantially increasing the number of kidneys available for
allograft survival.3 However, the field continues to face transplant by 2030.6 These efforts were initially hampered
important challenges. The vast majority of the more than by the global novel coronavirus disease 2019 (COVID-19)
700,000 persons in the United States with kidney failure pandemic, although rates of both deceased donor trans-
will not have an opportunity to receive a transplant due to plantation and LDKT subsequently recovered to prepan-
limitations in organ supply. Those who receive a transplant demic levels.7,8,9
often ultimately return to dialysis due to limited long-term To assess the knowledge gaps amenable to research
graft survival resulting from chronic allograft nephropathy, support aimed at advancing the practice and outcomes of
recurrence of native disease, medication nonadherence, or kidney transplantation, the National Kidney Foundation
other causes.2,4 Efforts to expand the deceased donor organ (NKF) convened an expert panel. The panel was charged
supply have increased the use of kidneys from older and with developing a research agenda to facilitate access to
other nonstandard donors to some extent, yet many kidney transplantation for all patients who can benefit,

AJKD Vol 78 | Iss 3 | September 2021 319


Lentine et al

A Candidate Age at Listing, by Listing Years B Candidate Race/Ethnicity, by Listing Years


100% 100%

80% 80%

60% 60%

40% 40%

20% 20%

0% 0%
2000-2005 2006-2010 2011-2016 2017-2020 2000-2005 2006-2010 2011-2016 2017-2020
<=18 19 to 30 31 to 44 45 to 59 >=60 White Black Hispanic Other

C Candidate BMI at Listing, by Listing Years D Candidate Cause of Kidney Failure, by Listing Years
100% 100%

80% 80%

60% 60%

40% 40%

20% 20%

0% 0%
2000-2005 2006-2010 2011-2016 2017-2020 2000-2005 2006-2010 2011-2016 2017-2020
<18.5 18.5 to <25 25 to <30 30 to <35 >=35 Hypertension Diabetes mellitus
Glomerulonephritis Polycystic kidney disease
Other/Unknown

Figure 1. Changing characteristics of the US kidney transplant waiting list. Prepared from Scientific Registry of Transplant Recip-
ients data, 2010-2020. Age is shown in years; body mass index (BMI) is shown in kg/m2.

support the goal of “one transplant for life” for organ Priorities for Research Innovation
recipients, and reduce/eliminate racial and ethnic dis-
parities in transplant access and outcomes. Participants Priority 1: Expand Opportunities for Safe Living
included nephrologists, surgeons, organ procurement Donation and Access to LDKT
organization (OPO) leaders, NKF leadership, patients, and Living kidney donors can be related, unrelated, nondi-
other interested parties. Consensus-building was con- rected, or participants in donor exchange (kidney paired
ducted through email, conference calls, and an in-person donation [KPD]) programs. Benefits of LDKT over
roundtable meeting December 13, 2019, in New York deceased donor kidney transplantation include faster
City. Through facilitated discussions, the panel developed 7 access to transplant and superior patient and allograft
priorities for research innovations. The in-person meeting survival at the lowest cost to the health care system.1 In
occurred shortly before the COVID-19 pandemic, and the United States, the number of LDKT declined from
recommendations have been updated in the context of the 6,000 in 2004 to approximately 5,500 per year through
latest challenges facing the field. 20173; 2018-2019 showed a promising trend of

320 AJKD Vol 78 | Iss 3 | September 2021


Lentine et al

1200
Paired Donation Non-Directed Donation
1000

Counts of LDKT 800

600

400

200

0
'00 '01 '02 '03 '04 '05 '06 '07 '08 '09 '10 '11 '12 '13 '14 '15 '16 '17 '18 '19
Year

Figure 2. Counts of kidney paired donation and nondirected donation over time. Prepared from Scientific Registry of Transplant Re-
cipients data, 2000-2019. Abbreviation: LDKT, living donor kidney transplant.

an increase to nearly 6,900 LDKT, although growth was Other strategies for increasing LDKT include educa-
stalled in 2020 by the temporary disruption of nearly all tional interventions, public campaigns, evaluation effi-
LDKT activity at the start of the COVID-19 pandemic.9 ciencies, and donor cost mitigation efforts.16 In a scoping
Barriers to LDKT include the transplant candidate’s review, Barnieh et al identified 26 studies reporting stra-
ability to identify a willing and appropriate donor, tegies to increase LDKT.17 Patient education was the only
concerns for donor health and follow-up, and economic strategy empirically assessed by randomized clinical trials,
barriers to donation including out-of-pocket expenses and only 2 (involving home-based education of transplant
and lost wages.10 candidates and members of their social network) found
Development of KPD programs for patients with willing significant increases in LDKT and living donor evalua-
but biologically incompatible donors has been an impor- tions.18,19 Despite the urgent need to increase access to
tant advance in expanding opportunities for LDKT.11 Using LDKT, only 6 quasi-experimental and 13 observational
algorithms to exchange kidneys among 2 or more studies assessed diverse strategies such as education, organ
donor–recipient pairs, KPD programs create compatible donation awareness campaigns, advocacy (training a
combinations that address blood group and donor-specific donor champion), efficiency (web-based screening, crea-
alloreactivity incompatibilities. Nondirected donors (ie, tion of interdisciplinary teams), removal of disincentives
donors who do not have an identified recipient) provide (leave policies, tax benefits), and institution of KPD pro-
the unique potential to expand the donor pool through grams. The only quasi-experimental study in the system-
chains of kidney exchanges.12 KPD is the fastest growing atic review that identified an increase in LDKT was a
LDKT modality and currently comprises approximately structured educational program for potential recipients and
16% of LDKT per year (Fig 2).3 However, KPD is under- their families.20 Further, well-designed evaluation trials are
used due to cost, complexity, patient and physician needed to identify successful interventions for increasing
acceptance, and degree of allosensitization. If all centers LDKT, especially for populations with reduced LDKT
used KPD at the rate of high-performing centers, it is access.
estimated that another 1,000 transplants could be per- Recently, a growing body of evidence has quantified
formed annually.13 Entering compatible pairs into KPD is and continues to improve estimates of the potential risks of
another suggestion to further increase the pool of potential living donation for donors, such as a tool using donor
matches and provide benefits such as better balance candidate demographic and health characteristics to esti-
in kidney mass/function of exchanged donors, and mate (in the absence of donation) the 15-year and lifetime
improved degree of tissue matching (ie, reduced eplet risk of kidney failure (https://www.ckdpc.org/tools.
mismatches) to lower long-term rejection risk.14 Addi- html).21 Additional research is required to validate and
tional innovations in some KPD programs include increase the precision of such tools22 and to incorporate
“advanced donation,” wherein a donor gives to the KPD emerging risk factors (eg, apolipoprotein L1 [APOL1] as a
pool in exchange for a voucher for a donation to their genetic risk marker).23 Unfortunately, methods for effec-
intended recipient at a later time if and when needed, tively communicating risk information to potential donors
designed to overcome “chronological incompatibility.”15 have been poorly studied. Even though interventions to

AJKD Vol 78 | Iss 3 | September 2021 321


Lentine et al

Educaon & Outreach Removal of Disincenves


Target audiences Uncompensated costs for donors
• Paents with kidney failure • Travel
• Potenal living donors • Medicaons
• Social networks • Lost me from work
• General public • Dependent Care
Research needed to opmize Research needs
• Frequency • Develop and assess mechanisms
• Content for achieving cost neutrality
• Delivery modalies

Evaluaon & Process Efficiency Improving Safety & Defensibility


Complex donor evaluaon process of Donor Selecon
• Delays may discourage donors Transparency
• Delays may lead to recipient • Incomplete risk assessment and
starng dialysis before transplant disclosure may reduce trust of the
Research needs public and potenal donors
• Develop standards to address Research needed to opmize
modifiable process delays • Scope and precision of long-term
• Remove logiscal barriers to KPD risk predicon
parcipaon • Methods for risk communicaon

Figure 3. Focal points for future research to define effective strategies for increasing access to living donor kidney transplant.
Adapted from Lentine and Mandelbrot10 with permission of the American Society of Nephrology (ASN). Original content ©2017
ASN. Abbreviation: KPD, kidney paired donation.

improve transparent communication of risks and long- when programs are scaled and to identify unexpected
term donor safety are not primarily directed at increasing barriers that may arise with wider implementation.
LDKT, these efforts are critical to sustaining trust in LDKT
and for informing evidence-based education and donor Recommendations
selection practices.24 Efforts to develop registries tracking
The panel agreed with recent suggestions that high-priority
long-term living donor outcomes in diverse populations
research to increase access to LDKT should include devel-
are also needed.25,26
oping, accessing and expanding strategies (Fig 3)10 to:
Attention to improving equity in LDKT access for Black,
Hispanic, and other non-White patients, as well as those • Improve education and outreach to kidney patients about
with lower levels of socioeconomic status and health lit- effective strategies for potential donor identification;
eracy, is a vital priority.27 Although removing financial • Reduce financial disincentives to donation;
disincentives is broadly accepted as necessary to improve • Improve the efficiency of donor evaluations and predo-
equity in donation and access to LDKT, evidence demon- nation processes, with particular attention to removing
strating the impact of programs targeting financial disincentives and logistical barriers to KPD participation
neutrality on donation rates has been limited to date.28 for programs and patients;
Addressing financial burdens to donors is particularly vi- • Optimize donor risk assessment and long-term follow-
tal, as a pandemic-related recession may challenge op- up; and
portunities for living donation beyond the end of the • Identify and implement methods to improve risk
public health emergency. Technology may enable efficient communication, transparency, and shared decision-
education delivery, but evidence is needed to determine if making in the donor candidate evaluation.
e-learning, telehealth, or telephone-based LDKT education
programs are as effective as in-person education. Further Priority 2: Improve Waitlist Access, Management,
research is also needed to improve the efficiency of donor and Pretransplant Readiness
evaluations and to generate evidence-based standards Among the nearly 95,000 patients waiting for kidney or
to address modifiable process delays.29,30 Dissemination combined kidney/pancreas transplant in March 2021, 43%
studies are also necessary to assess whether effects persist had an inactive status.31 Thus, despite being approved for,

322 AJKD Vol 78 | Iss 3 | September 2021


Lentine et al

and presumably in need of, a kidney transplant, these sufficiently to allow transplant. Further work is needed to
patients will not appear on a match run for organ offers. identify the best assessment tools and successful in-
Patients are inactive for a variety of reasons (eg, current terventions to facilitate access to transplantation and
medical illness, psychosocial barriers, preserved kidney improve posttransplant outcomes for frail kidney patients.
function that meets listing criteria but does not warrant The COVID-19 pandemic created a new emphasis on
kidney replacement therapy). Patients who are initially remote assessments, including of candidate physical
listed as inactive have twice the risk of waitlist mortality functioning at evaluation and while awaiting a transplant
and 32% lower rates of eventual transplantation than pa- offer.9
tients initially listed as active.32 The prevalence of inactive
status on the waiting list is higher among racial and ethnic Recommendations
minority populations. One study of national US waitlist • Optimize strategies to eliminate barriers to waitlist
records found that, among patients who were inactive, acceptance, including management of obesity, frailty,
Black patients were 19% less likely to convert to active and deficiencies in adequate psychosocial support,
status and Hispanic patients were 27% less likely than their grounded in transparent communication of eligibility
White counterparts.33 This disparity directly impacts access criteria to patients and referring clinicians.
to transplantation and overall mortality. • Develop and implement educational interventions that
The process of referral and evaluation for transplant is assist all patients, particularly members of racial and
complex and often difficult to navigate. Patients ethnic minorities, to successfully reach the waitlist
must complete rigorous testing and examinations to ensure expeditiously.
that there are no medical or surgical contraindications to • Implement scalable and generalizable information tech-
transplantation. Racial and socioeconomic disparities in nology solutions to support waitlist access and mainte-
completing the evaluation are well documented and persist nance of transplant readiness.
even after controlling for other social determinants of
health.34 Time- and resource-intensive interventions (eg,
patient navigators, individually tailored educational pro- Priority 3: Decrease Organ Discard by Expanding
grams, coaching) have increased the proportion of patients Clinician and Patient Organ Acceptance
who are successfully waitlisted in single-center trials.35 Long waiting times for kidney transplantation create a
The panel agreed that there is an urgent need to strong imperative to make the best use of donated organs.
develop and disseminate best practices for transplant Nearly 10% of individuals with a previously declined
centers to reduce inactivation status on their waiting lists. organ offer die on the waitlist without a transplant, and
Kataria et al reported that frequent programmatic review another 20% are removed from the waitlist without a
allowed 18% of inactive patients to be made active, transplant. The United States has the highest rate of organ
although this also led to delisting of 40% of inactive pa- recovery without transplantation (nonutilization or
tients at their center.36 The panel considered research “discard”), at nearly 20%.44 The federal government has
opportunities that target specific barriers with effective in- focused national attention on increased organ recovery
terventions. For example, patients with a body mass index and reduced organ discard rates as the first objective of
exceeding center acceptance thresholds can be referred for the Advancing American Kidney Health executive order’s
nutritional interventions and possible bariatric surgery.37,38 increasing kidney transplant goal.6 There are opportu-
Bariatric procedures have been shown to achieve durable nities to increase the number of deceased donor kidney
weight loss and increase transplant access for appropriate transplants by measuring and maximizing the potential
candidates.39,40 A systematic review/meta-analysis including pool of deceased donor kidneys and minimizing the
288 patients with advanced kidney disease demonstrated number of organ nonutilizations.45,46 Expanding the pool
that 50% lost sufficient weight for transplant listing.40 of accepted organs will likely require multifaceted efforts
However, access to bariatric surgery remains limited for that address barriers at the levels of patients, transplant
the population with advanced kidney disease. centers, and OPOs.44 Furthermore, researchers and
Frailty is another frequent barrier to kidney trans- funding organizations should invest in developing new
plantation, such that frail patients are half as likely as technologies to optimize early organ function, assess
nonfrail counterparts to be listed for transplant.41-43 transplantability of higher-risk or damaged organs, and
Among those who are listed and receive transplants, the foster the potential to repair injured kidneys prior to
approximately 20% of recipients who are determined to transplant.
be frail at transplant are twice as likely to die after There is abundant evidence that many deceased donor
transplant as nonfrail recipients. Current data suggest that kidneys that could benefit some patients are not utilized
objective frailty scoring systems, rather than subjective or never recovered. For example, although the number of
physician assessments, are more accurate and should be patients aged 65 years and older continues to rise (20.5%
used at the time of evaluation.42,43 For frail patients, a of additions to the waiting list in 2020), the average age of
structured program of “prehabilitation” and physical deceased kidney donors has not increased in parallel. A
therapy may help improve physical functioning study comparing deceased donor transplant practices in the

AJKD Vol 78 | Iss 3 | September 2021 323


Lentine et al

Discard Proportion DGF Freedom Graft Survival (5 yr)


100%
88.7% 86.9%
90%
84.0% 81.1%
89.1% 76.6%
80%
70.2%
70%
75.9%
63.5% 67.1%
69.9%
66.9% 62.4%
60%

50% 53.8%

40%
32.8%

30%

20%
19.2%
10%
2.5%
9.5%
0%

Kidney Donor Profile Index

Figure 4. Deceased donor kidneys: use and outcomes. The discard rate rises more rapidly with higher Kidney Donor Profile Index
compared to the graft failure and delayed graft function (DGF) rates. Prepared from Scientific Registry of Transplant Recipients data,
2010-2020.

United States versus France in 2004-2014 found that the (eg, in the presence of significant nephrolithiasis or surgical
mean donor age of kidneys transplanted in the United injury), many of these decisions appear to be based on
States was substantially lower, 36.2 versus 50.9 years.47 inaccurate data, including procurement biopsies.49,50 Evi-
Donor age and mean Kidney Donor Risk Index (KDRI) dence shows that kidneys are more likely be discarded on
steadily rose over time in France, while donor age and the weekend and that organs that are accepted and suc-
KDRI of kidneys transplanted in the United States showed cessfully transplanted on the weekend are declined many
little change, suggesting significant untapped donor po- more times before eventually being accepted. Differences in
tential.47 Studies of US registry data demonstrate a strong organ acceptance on a weekend versus a weekday suggests
relationship between organ quality scores and discard that factors beyond organ quality and patient issues influ-
rates, such that more than 45% of kidneys with Kidney ence the willingness of transplant centers to accept an or-
Donor Profile Index (KPDI) scores >80% are discarded gan.51 Frequently cited reasons for organ offer declines
(Fig 4).45 Despite higher rates of graft failure, high-KDPI include the desire to wait for a “better organ offer” for
organs can provide survival and quality-of-life benefits patients at the top of the waitlist, as well as concerns about
over maintenance dialysis in appropriate patients. A program outcomes.49,51 Taken together, these findings
national registry study found that transplant with high-KDPI imply a substantial opportunity to transplant many currently
kidneys (defined as KDPI >70) was associated with discarded kidneys, particularly if the offers are targeted to
increased short-term death risk compared to remaining on willing patients and centers.
dialysis for 6 additional months. However, these organs Many programs report kidney nonutilization due to
improved long-term survival compared to waiting on dial- concerns for posttransplant outcomes that, if markedly
ysis for a higher-quality organ among those who were worse than those at peer centers, may lead to censure by
older, had diabetes, or were living in areas with prolonged the Organ Procurement and Transplantation Network/
waiting times.48 The highest-risk organs (KDPI 91-100) United Network for Organ Sharing (OPTN/UNOS), loss of
appeared to benefit patients older than age 50 years at private payer contracts, and decreased transplant volume.52
centers with median waiting times longer than 33 Additionally, managing delayed graft function (DGF;
months.48 typically defined as need for dialysis in the first week after
As another example, unilateral discard (in which only 1 transplant) creates additional burdens and costs for pro-
of 2 recovered kidneys is transplanted) is a relatively com- grams. These costs pose a significant financial disincentive
mon occurrence that calls into question the reliability of for accepting lower-quality kidneys.53 Care of transplant
organ evaluation. While some discordant discard is expected recipients who need dialysis during weeks 2-12

324 AJKD Vol 78 | Iss 3 | September 2021


Lentine et al

posttransplant requires increased resources and costs.54 benefits of accepting kidneys at risk for discard (KARD),
Yet, over the life of the organ, there are significant clin- including organs from donors with older age, lower
ical and economic benefits of transplantation with quality scores, acute kidney injury, behaviors associated
nonstandard, higher-risk organs.1 Developing alternative with viral infections, or certain known viral infections.
payment methodologies to eliminate financial barriers to • Create and evaluate communication strategies to effec-
nonstandard organ acceptance is vital to achieve the goal of tively educate patients and clinicians about organ offers
increasing organ acceptance.55 and promote shared decision-making at listing and
Patients and some clinicians may also be dissuaded from throughout the experience on the waiting list.
accepting kidneys due to perceived stigma associated with • Incorporate additional definitions of success (incremen-
donors with a history of injection drug use or sex work, tal survival, quality of life improvement) into models of
despite the very small probabilistic estimates of viral transplant acceptance.
transmission to recipients.56 Kidneys from infection risk
donors (IRDs) defined by US Public Health Services (PHS)
criteria continue to be discarded at elevated rates, but Priority 4: Decrease Organ Discard With Use of
provide significant benefit over dialysis. IRD kidneys are Novel Assessment Technologies and Real-Time
often high-quality organs from young donors with Decision Support
excellent long-term survival, which confer minimal Given the growing complexity of organ acceptance to
infection transmission risk in the era of universal nucleic optimize competing concerns for graft failure versus death
acid testing. In December 2020, the OPTN revised policy on the waiting list, logistical complexity exacerbated by
related to IRD organs to align with the 2020 updated US geographically broader organ sharing, and clinician con-
PHS Guideline, including replacing the requirement to cerns for patient and program outcomes, decision support
obtain specific “informed consent” with a requirement systems may promote KARD acceptance. Despite wide-
that transplant clinicians inform intended recipients when spread use and integration into organ allocation, the KDPI
the donor has any risk criteria, combined with increased is a first-generation tool to assess kidney quality that has
posttransplant monitoring of all recipients.57 The impact of modest ability to predict graft survival and carries a sig-
this policy change on acceptance of organs from IRD do- nificant chance of misclassifying risk, which may increase
nors by patients and clinicians, and on posttransplant the likelihood of organ discard.63 Investigators should seek
outcomes, should be closely monitored. to develop tools for transplant clinicians to objectively
The introduction of direct-acting antiviral (DAA) medi- assess the benefits of specific kidney offers for individual
cations that achieve sustained viral response (cure) rates patients, accounting for logistical realities during time-
above 95% with few side effects has expanded opportunities sensitive and complex decision-making. These tools
to safely use organs from donors with hepatitis C viremia might incorporate machine learning, artificial intelligence
(HCV) in uninfected recipients, which, when coupled with (AI), and other advanced analytics to integrate the complex
DAA posttransplant therapy, results in excellent outcomes.58 clinical information available about donors and candidates
Nonetheless, up to 40% of US transplant programs remain to match organs to appropriate recipients. Efficient in-
reluctant to incorporate transplants from donors with HCV terfaces are needed with OPTN/UNOS systems to provide
into their practice due to barriers including insurance data in real time without additional data entry. For
coverage concerns, cost, and perceived risk of transmitting example, integrated interpretation of complex serum
resistant infection.59 The COVID-19 pandemic created an creatinine trajectories, renal pathology images, or machine
urgent need to develop tools for rapid and reliable screening perfusion parameters such as resistance and flow patterns
of donors for a new viral infection.60 As the prevalence of could be facilitated by AI or advanced modeling tools.
individuals with recovered COVID-19 increases in the Clinicians and scientists have proposed multiple expla-
population, determining if and when organs from donors nations for high kidney discard rates in the United States;
with recovered COVID-19 can be safely used is a critical however, the primary code used for organ decline is organ
research priority.61,62 quality, often related to biopsy results.45 Nearly half of
Importantly, metrics of transplant success have focused deceased donor kidneys in the United States undergo post-
largely on patient and graft survival. However, for some recovery biopsy to evaluate “quality,”50 despite nearly uni-
patients—especially the elderly—the most important form evidence refuting the benefit of these biopsies, which
benefits of transplantation may relate to other domains add to cost and cold ischemia time. A study comparing
such as quality of life or greater social participation. allocation in the United States versus France and Bel-
Freedom from dialysis, even for a shorter period, may be gium—where kidneys are not routinely biopsied during
highly valuable for patients and families. allocation—reported no predictive value of biopsies over
what is routinely available from donor medical records.64
Recommendations The reliability of kidney biopsy is further complicated by
• Develop and evaluate patient- and clinician-facing educa- the limited availability of trained renal pathologists and the
tional materials to foster understanding of the potential difficulty of interpreting frozen sections due to artifact.

AJKD Vol 78 | Iss 3 | September 2021 325


Lentine et al

Telepathology allows remote review and image interpreta- the organ and, potentially, the opportunity to improve its
tion, while whole-slide interpretation using AI systems ap- quality. Normothermic perfusion of kidney allografts can
pears promising to improve reliability of biopsy reports. permit assessment of graft function in an ex vivo setting.
Accurate biopsy data utilizing AI solutions could facilitate Successful transplant of a kidney with severe acute kidney
more rapid placement of organs, as the community appears injury from rhabdomyolysis following effective warm
not yet ready to significantly limit biopsies, as some have perfusion has been reported.70 A prospective trial of
proposed.65,66 A pilot trial to assess the feasibility of surgeon normothermic perfusion is currently under way to assess
willingness to defer access to biopsy information at the time impacts on graft utilization and outcome.71
of offer acceptance is under way.67 Alternatively, hypothermic oxygenated perfusion can be
performed. A small cohort study comparing extended-
Recommendations criteria donor kidneys with matched historical controls
• Identify tissue, urine, or circulating biomarker tools to (15 perfused kidneys vs 30 controls) failed to demonstrate
better predict kidney graft survival, and inform organ significant benefits in rates of DGF or graft failure.
acceptance decisions. Ongoing trials in Europe are underway to assess the value
• Define appropriate use of and approach to procurement of hypothermic oxygenated perfusion in kidney allografts
biopsies by: from extended-criteria donors.72 However, more work is
> Assessing the impact of organ selection without bi- needed to determine which perfusion techniques lead to
opsy on patient-centered outcomes (discard, renal optimal outcomes in different clinical contexts.
function, graft survival, patient survival). Finally, novel methods to improve marginal kidney
> Evaluating the accuracy of whole-slide/multilevel bi- function during ex vivo perfusion using pharmacologic,
opsy review including AI-supported reading. cellular, and genetic therapies are currently being
• Develop AI systems to integrate complex, multifaceted explored.73 These therapies are designed to mitigate
information including kidney biopsy data, novel bio- ischemic reperfusion injury and diminish alloimmune
markers, and other donor risk information to: activation and organ damage. Ex vivo treatment offers
> Generate graft survival predictions tailored for patient significant potential advantages. First, the therapy can be
clinical profiles. targeted directly to the organ without impacting the po-
> Reduce inter-clinician variability in organ acceptance. tential recipient. Second, the intervention is limited to the
kidney and will not impact other organs recovered from
the same donor. The infusion of mesenchymal stromal
Priority 5: Develop and Implement Strategies to cells has been evaluated as a method to decrease allor-
Preserve, Resuscitate, or Evaluate Kidney eactivity and induce tolerance. Mesenchymal stromal cells
Allografts Before Implantation can hone in on injured cells and promote recovery through
Technology for preservation of kidney allografts has not the release of growth factors and cytokines.74 Preclinical
advanced substantially over the past 2 decades. Static cold studies suggest that mesenchymal stromal cell infusion
storage in electrolyte-rich preservation solutions can allow during normothermic perfusion results in decreased in-
successful transplantation of kidneys with prolonged cold flammatory cytokines and markers of ischemia-reperfusion
ischemic times (CITs) even exceeding 36 hours. However, injury. Gene therapy, such as methods that have been
organs preserved with static cold storage alone for widely used in altering T-cell receptor specificity, has been
extended periods have high rates of DGF. Hypothermic suggested as a method to promote tolerance, limit the
pulsatile machine perfusion has been demonstrated to detrimental effects of ischemic reperfusion, and modify
reduce the incidence of DGF and costs of the transplant host–organ interaction in xenotransplantation. The only
hospitalization. In a randomized trial of 752 paired kid- clinical trials to date of therapy administration during
neys, machine perfusion resulted in significantly reduced kidney perfusion have utilized pharmacologic agents, and
DGF in transplants with CIT less than 10 hours (6% vs did not yield significant reductions in DGF rates.
28%; P < 0.002) but did not significantly decrease DGF in
the context of longer CIT.68 Pulsatile perfusion appears to Recommendations
have benefits for both standard-criteria and higher-risk • Investigate optimal preservation strategies (including
kidneys. A retrospective analysis of US registry data innovative perfusion technologies) to decrease DGF rates
found reduced DGF rates after pulsatile perfusion of kid- and improve organ utilization.
neys from both standard and higher-risk donors.69 • Evaluate novel techniques to monitor transplants during
Innovations have extended the benefits of perfusion preservation, improve the accuracy of quality assess-
technology from preservation alone to organ assessment, ments, and optimize utilization.
improvement, and resuscitation. While standard hypo- • Examine ex vivo interventions directed at improving
thermic pulsatile perfusion flow and resistance parameters allograft function, reducing ischemia-reperfusion injury,
have been correlated with graft outcomes, innovations in and extending the life of donor allografts through im-
perfusion technology promise greater information about mune regulation.

326 AJKD Vol 78 | Iss 3 | September 2021


Lentine et al

Box 1. Summary of Research Priorities to Advance Kidney Transplant Access and Outcomes

Priority 1: Expand Opportunities for Safe Living Donation and Access to LDKT
Challenges Recommended Research Targets
• Transplant candidates have varying abilities to • Improve education and outreach to kidney patients about
share their need and identify potential living effective strategies for potential donor identification
donors • Reduce financial disincentives to donation
• Lack of information and disincentives may deter • Improve the efficiency of donor evaluations and predonation
willing, healthy persons from donation: processes, with particular attention to removing disincentives
• Gaps in ensuring donor follow-up and and logistical barriers to KPD participation for programs and
complete risk assessment patients
• Economic barriers, including out-of-pocket • Optimize donor risk assessment and long-term follow-up
expenses and lost wages • Identify and implement methods to improve risk communica-
tion, transparency, and shared decision-making in the donor
candidate evaluation
Priority 2: Improve Waitlist Access, Management, and Pretransplant Readiness
Challenges Recommended Research Targets
• Racial and ethnic minority populations face dis- • Optimize strategies to eliminate barriers to waitlist accep-
parities in access to transplantation and overall tance including management of obesity, frailty, and de-
waitlist mortality ficiencies in adequate psychosocial support, grounded in
• Weight-loss methods to improve BMI, like transparent communication of eligibility criteria to patients and
nutritional interventions and bariatric surgery, referring clinicians
are not accessible for many obese patients • Develop and implement educational interventions that assist
• Structured or standardized “prehabilitation” and all patients, particularly members of racial and ethnic minor-
physical therapy programs to improve physical ities, to successfully reach the waitlist expeditiously
function prior to transplant are not accessible for • Implement scalable and generalizable information technology
many frail patients solutions to support waitlist access and maintenance of
• Referral and evaluation are complex, multi-step transplant readiness
processes
Priority 3: Decrease Organ Discard by Expanding Clinician and Patient Organ Acceptance
Challenges Recommended Research Targets
• On average, 20% of kidneys are discarded after • Develop and evaluate patient- and clinician-facing educational
recovery, and this proportion rises sharply for materials to foster understanding of the potential benefits of
nonstandard organs accepting a KARD, including organs from donors with older
• Patient and clinician perceptions and education age, lower quality scores, AKI, behaviors associated with viral
limit acceptance of organs from donors with high infections, or certain known viral infections
KDPI, increased risk of viral infection, or some • Create and evaluate communication strategies to effectively
known viral infections educate patients and clinicians about organ offers and pro-
• Metrics of transplant success have focused mote shared decision-making at listing and throughout the
largely on patient and graft survival, and not experience onf the waiting list
incorporated QoL • Incorporate additional definitions of success (QoL improve-
ment) into models of transplant acceptance
Priority 4: Decrease Organ Discard Through Use of Novel Assessment Technologies and Real-Time Decision
Support
Challenges Recommended Research Targets
• Despite integration into organ allocation, the • Identify tissue, urine, or circulating biomarkers tools to better
KDPI has modest ability to predict graft survival predict kidney graft survival and inform organ acceptance
and may misclassify true organ quality decisions
• Biopsied kidneys are 3-times more likely to be • Define appropriate use of and approach to procurement bi-
discarded despite: opsies by:
> Marked variation in indications for biopsy, > Assessing the impact of organ selection without biopsy
technique, and expertise in interpretation on patient-centered outcomes (discard, kidney function,
> Limited reliability of standard frozen section graft survival, patient survival)
biopsy and uncertain prognostic value > Evaluating the accuracy of whole slide/multilevel review
including AI-supported reading
(Continued)

AJKD Vol 78 | Iss 3 | September 2021 327


Lentine et al

(Cont'd).
• Develop AI systems to integrate complex, multifaceted infor-
mation including kidney biopsy data, novel biomarkers and
other donor risk information to:
> Generate graft survival predictions tailored for patient
clinical profiles
> Reduce inter-clinician variability in organ acceptance
Priority 5: Develop Strategies to Preserve, Resuscitate, or Evaluate Kidney Allografts Before Implantation
Challenges Recommended Research Targets
• Technology for preservation of kidney allografts • Investigate optimal preservation strategies (including innova-
has not advanced substantially over the past 2 tive perfusion technologies) to decrease DGF rates and
decades improve organ utilization
• Innovations in organ perfusion, assessment, and • Evaluate novel techniques to monitor transplants during
resuscitation are infrequently used preservation, improve the accuracy of quality assessments,
and optimize utilization
• Examine ex vivo interventions directed at improving allograft
function, reducing IRI, and extending the life of donor allo-
grafts through immune regulation
Priority 6: Sustaining One Transplant for Life
Challenges Recommended Research Targets
• Potent IS has substantially reduced acute cellular • Develop personalized strategies for posttransplant IS man-
and humoral rejection, but long-term graft survival agement incorporating novel assessment tools such as
remains limited GWAS data for donors and recipients and novel posttrans-
• IS is associated with complications including plant monitoring tools
infection, malignancy, glucose intolerance, • Assess the impact of tolerance induction strategies on patient
increased costs of care, and decreased QoL and graft survival, QoL, and development of IS complications
Priority 7: Optimize Kidney Transplantation for Pediatric Patients
Challenges Recommended Research Targets
• Pediatric kidney failure is associated with multi- • Evaluate strategies to improve coordination of care transitions
systemic sequelae including impacts on between pediatric and adult transplant clinicians
cognition • Identify approaches to improve medication adherence and
• The highest risk period for graft loss among pe- reduce early graft loss while protecting the neurocognitive
diatric transplant patients occurs in late teenage/ development of children and adolescents
early adulthood years, prior to complete neuro- • Optimize organ allocation and IS management to reduce the
development and concomitant with transition incidence and complications of viral diseases in pediatric
from pediatric to adult care teams recipients
• Repeat transplantation of children with allograft
failure is complicated by higher rates of
allosensitization
• Children face high risk of viral-driven malignancy
from donor-derived viral infections
Abbreviations: AI, artificial intelligence; AKI, acute kidney injury; BMI, body mass index; GFR, glomerular filtration rate; GWAS, genome-wide association study; KARD,
kidney at risk for discard; KDPI, kidney donor profile index; KPD, kidney paired donation; IRI, ischemic reperfusion injury; IS, immunosuppression; LDKT, living-donor
kidney transplantation; QoL, quality of life.

Priority 6: Sustain One Transplant for Life mammalian target of rapamycin inhibitors (mTORis) may
Although contemporary tissue-typing techniques have limit immunosuppression-associated toxicities.76,77 Howev-
essentially eliminated hyperacute rejection, and potent er, novel strategies are needed to achieve the goal of one
immunosuppression has substantially reduced acute cellular transplant for life.
and humoral rejection, long-term graft survival remains The development of personalized and effective immu-
limited. Furthermore, immunosuppression is associated with nosuppression strategies requires a shift from phenotypic
complications including infection, malignancy, glucose (eg, demographics-based) assessments to genotypic
intolerance, and decreased quality of life.75 Personalization of assessment of patient characteristics.23 Accurate models
maintenance immunosuppression with appropriate applica- require the collection and analysis of biobanks from large
tions of steroid-sparing regimens, alternative pharmacologic and diverse patient populations. In the DeKAF and GEN03
agents (eg, extended-release tacrolimus), and judicious use of cohort studies, investigators utilized genome-wide

328 AJKD Vol 78 | Iss 3 | September 2021


Lentine et al

association studies (GWAS) to detect clinically significant patients may lead to structural brain abnormalities.89 While
differences in calcineurin inhibitor metabolism on the basis of significant cognitive impairment may not be present in most
a complex interaction of multiple genetic polymorphisms.78 children with chronic kidney disease, even subtle cognitive
These data demonstrate that the impact of recipient genetic impairments can pose barriers to understanding of and
variation is complex, differs by patient characteristics, and adherence to complex medication regimens required as part
extends beyond well-characterized genomic variation (eg, of caring for a transplant. The impact of these subtle
CYP34). Developing clinical application of genotypic data cognitive deficits cannot be overemphasized given that the
will benefit from systematic collection and analysis of donor highest-risk period for graft loss among pediatric transplant
and recipient genetic variations through GWAS, linked patients occurs in late teenage/early adulthood years when
directly with baseline clinical and outcomes information. critical periods of neurodevelopment are not yet complete,
Belatacept, a soluble fusion protein comprising the in parallel with periods of transition from pediatric to adult
modified extracellular domain of CTLA-4 fused to a portion care teams.90 Children also face high risk of viral-driven
(hinge-CH2-CH3 domains) of the Fc domain of a human malignancy from donor-derived viral infections.
immunoglobulin G1 antibody, has been evaluated in 2
prospective clinical trials as a novel alternative to oral Recommendations
immunosuppression.79 While initial studies suggested high • Evaluate strategies to improve coordination of care transi-
rates of rejection, pilot studies combining belatacept, tions between pediatric and adult transplant clinicians.
alemtuzumab induction, and slow tapering of calcineurin • Identify approaches to improve medication adherence
inhibitors have facilitated successful belatacept mono- and reduce early graft loss while protecting the neuro-
therapy in some patients. Alternative strategies under cognitive development of children and adolescents.
investigation include use of combined cellular and kidney • Optimize organ allocation and immunosuppresion
transplantation to induce a state of “tolerance” through management to reduce the incidence and complciations
chimerism, allowing tapering and elimination of mainte- of viral diseases in pediatric recipients.
nance immunosuppression.80,81 Incorporation of novel
biomarkers into posttransplant surveillance may help
Conclusions
identify which patients have lower immunologic risk and
could benefit from immunosuppression minimization to Kidney transplantation offers hope to thousands of patients
reduce complications without increasing risk of rejection.82 each year, but the goal of universal access to this treatment
remains elusive. Addressing the priorities outlined in this
Recommendations research agenda has the potential to transform kidney pa-
• Develop personalized strategies for posttransplant tient care by expanding opportunities for safe living
immunosuppression management, incorporating novel donation, improving waitlist access and transplant readi-
assessment tools such as GWAS data for donors and re- ness, maximizing use of available deceased donor organs,
cipients and novel posttransplant monitoring tools. and extending graft longevity. These priorities will frame
• Assess the impact of tolerance induction strategies on research and funding initiatives for the NKF and assist
patient and graft survival, quality of life, and develop- patients with chronic kidney disease in receiving and
ment of immunosuppression complications. nurturing the gift of transplant (Box 1).

Priority 7: Optimize Kidney Transplantation for Article Information


Pediatric Patients Authors’ Full Names and Academic Degrees: Krista L. Lentine,
Although patients younger than 20 years of age account MD, PhD, Stephen Pastan, MD, Sumit Mohan, MD, MPH, Peter P.
Reese, MD, MSCE, Alan Leichtman, MD, Francis L. Delmonico,
for less than 2% of the total North American patient MD, Gabriel M. Danovitch, MD, Christian P. Larsen, MD, PhD,
population treated by kidney replacement therapy, the Lyndsay Harshman, MD, MS, Alexander Wiseman, MD, Holly J.
prevalence of pediatric kidney failure has increased by 32% Kramer, MD, MPH, Joseph Vassalotti, MD, Jessica Joseph, MBA,
since 1990.83,84 Approximately 50% of all pediatric kidney Kevin Longino, MBA, Matthew Cooper, MD, and David A. Axelrod,
failure diagnoses are due to congenital, nonglomerular MD, MBA.
anomalies,85 and half of all pediatric patients with Authors’ Affiliations: Saint Louis University Center for Abdominal
congenital kidney disease will progress to a need for Transplantation, St Louis, MO (KLL); Department of Medicine,
Emory Transplant Center, Atlanta, GA (SP); Department of
transplantation during their lifetime.84 Pediatric kidney Medicine, Columbia University Medical Center (SM); National
failure is associated with an array of multisystemic Kidney Foundation (JV, JJ, KL); Department of Medicine, Icahn
sequelae, and the impact of kidney failure on cognition has School of Medicine at Mount Sinai (JV), New York, NY; Renal-
important implications for optimizing outcomes of trans- Electrolyte and Hypertension Division, University of Pennsylvania,
plantation in the pediatric population.86 Pediatric kidney Philadelphia, PA (PPR); Department of Medicine, Icahn School of
Medicine at Mount Sinai, New York, NY (AL); New England Organ
failure is associated with risk for academic underachieve- Bank, Wellesley Hills, MA (FLD); University of California, Los
ment87 and executive function deficits.88 Furthermore, Angeles, Los Angeles, CA (GMD); Department of Pediatrics (LH),
both the duration and severity of kidney disease in pediatric Department of Surgery (DAA), University of Iowa Transplant

AJKD Vol 78 | Iss 3 | September 2021 329


Lentine et al

Institute, Iowa City, IA (LH); Department of Medicine, Centura 2. Alhamad T, Axelrod D, Lentine KL. The epidemiology, out-
Health-Porter Adventist Hospital, Aurora, CO (AW); Department of comes, and costs of contemporary kidney transplantation.
Medicine, Loyola University, Chicago, IL (HJK); Department of In: Himmelfarb J, Ikizler TA, eds. Chronic Kidney Disease,
Surgery, Medstar Georgetown Transplant Institute, Washington, Dialysis, and Transplantation: A Companion to Brenner
DC (MC) and Department of Surgery, Emory Transplant Center, and Rector’s the Kidney. 4th ed. Elsevier; 2019:chapter
Atlanta, GA (CPL). 34.
Address for Correspondence: Krista L. Lentine, MD, PhD, Saint 3. Hart A, Lentine KL, Smith JM, et al. OPTN/SRTR 2019 Annual
Louis University Center for Abdominal Transplantation, 1201 S Data Report: kidney. Am J Transplant. 2021;21(suppl 2):21-
Grand Blvd, St Louis, MO, 63104. Email: krista.lentine@health.slu. 137.
edu 4. Alhamad T, Lubetzky M, Lentine KL, et al. Kidney recipients with
Support: The Transplant Research Roundtable was supported by allograft failure, transition of kidney care (KRAFT): a survey of
the National Kidney Foundation. No additional support was obtained. contemporary practices of transplant providers. Am J Trans-
Financial Disclosure: Dr Lentine reports receiving consulting fees plant. 2021. Published online February 8, 2021. doi:10.1111/
from CareDx, speaker honoraria from Sanofi, and research support ajt.16523.
from the National Institutes of Health (NIH; U01DK116042, 5. Purnell TS, Luo X, Cooper LA, et al. Association of race and
R01DK120551, and R01DK125018) and the Mid-America ethnicity with live donor kidney transplantation in the United
Transplant Foundation; she is a senior scientist of the Scientific States from 1995 to 2014. JAMA. 2018;319(1):49-61.
Registry of Transplant Recipients. Dr Pastan reports membership 6. Lentine KL, Mannon RB. The Advancing American Kidney
on the Board of Directors for the NKF and the Forum of End-Stage Health (AAKH) Executive Order: promise and caveats for
Renal Disease (ESRD) Networks, and is a Chairman of the local expanding access to kidney transplantation. Kidney360.
Board of IPRO/ESRD Network 6. Dr Mohan reports grant support 2020;1(6):557-560.
from NIH (R01DK126739, U01DK114893, and R01MD014161) 7. Pereira MR, Mohan S, Cohen DJ, et al. COVID-19 in
and Angion Biomedica and personal fees from Kidney International solid organ transplant recipients: Initial report from the
Reports and Angio Biomedica. Dr Reese reports having received US epicenter. Am J Transplant. 2020;20(7):1800-1808.
research support to the University of Pennsylvania from Merck, 8. Lentine KL, Vest LS, Schnitzler MA, et al. Survey of US
AbbVie, and Gilead to investigate transplantation of hepatitis C living kidney donation and transplantation practices in the
virus–infected organs into uninfected recipients followed by antiviral COVID-19 era. Kidney Int Rep. 2020;5(11):1894-1905.
treatment; serves as AJKD Associate Editor; and is an unpaid 9. Lentine KL, Mannon RB, Josephson MA. Practicing with
consultant to eGenesis, a company developing xenotransplantation uncertainty: kidney transplantation during the COVID-19
technology. Dr Leichtman is a consultant to Arbor Research pandemic. Am J Kidney Dis. 2021;77(5):777-785.
Collaborative for Health, a member of the governing board of Gift of 10. Lentine KL, Mandelbrot D. Moving from intuition to data:
Life Michigan, and a partner in Compass Integrated Care. Dr
building the evidence to support and increase living donor
Delmonico reports receiving salary from the New England Donor
kidney transplantation. Clin J Am Soc Nephrol. 2017;12(9):
Services in Massachusetts General Hospital. Dr Wiseman reports
1383-1385.
receiving consultant fees for Veloxis, CareDx, and Natera, and has
received speaking fees for Veloxis. Dr Kramer reports membership 11. Garg N, Gill J, Mandelbrot DA. Compatibility, kidney paired
on the NKF Board of Directors, serves as a consultant for Bayer donation, and incompatible living donor transplants. In:
Pharmaceuticals, and serves as AJKD Engagement Editor. Dr Lentine KL, Concepcion BP, Lerma EV, eds. Living Kidney
Vassalotti reports having served on a CKD advisory board for Donation: Best Practices in Evaluation, Care and Follow-up.
Renalytix AI, PLC. Ms Joseph and Mr Longino are employed by the Springer International; 2021:233-251.
NKF. Dr Cooper reports membership on the NKF Board of 12. Melcher ML, Leeser DB, Gritsch HA, et al. Chain trans-
Directors. Dr Axelrod reports receiving consultation fees from plantation: initial experience of a large multicenter program. Am
CareDx and Talaris and serves on the advisory board for Veloxis. J Transplant. 2012;12(9):2429-2436.
The remaining authors declare that they have no relevant financial 13. Massie AB, Gentry SE, Montgomery RA, Bingaman AA,
interests. Segev DL. Center-level utilization of kidney paired donation. Am
Acknowledgements: The authors thank Debra Taylor, MPH, for J Transplant. 2013;13(5):1317-1322.
assistance with manuscript preparation. The authors also thank 14. Gentry SE, Segev DL, Simmerling M, Montgomery RA.
Saint Louis University biostatisticians Huiling Xiao, MS, and Ruixin Expanding kidney paired donation through participation by
Li, MS, for assistance with SRTR data analysis and figure compatible pairs. Am J Transplant. 2007;7(10):2361-2370.
preparation. Data reported here have been supplied by the 15. Veale JL, Capron AM, Nassiri N, et al. Vouchers for future
Hennepin Healthcare Research Institute (HHRI) as the contractor kidney transplants to overcome “chronological incompatibility”
for the Scientific Registry of Transplant Recipients (SRTR). The between living donors and recipients. Transplantation.
interpretation and reporting of these data are the responsibility of 2017;101(9):2115-2119.
the authors and in no way should be seen as an official policy of 16. Hunt HF, Rodrigue JR, Dew MA, et al. Strategies for
or interpretation by the SRTR or the US Government.
increasing knowledge, communication, and access to living
Peer Review: Received April 17, 2021, following review and donor transplantation: an evidence review to inform patient
approval by the NKF Scientific Advisory Board (membership listed education. Curr Transplant Rep. 2018;5(1):27-44.
at https://www.kidney.org/about/sab) and the NKF Executive 17. Barnieh L, Collister D, Manns B, et al. A scoping review for
Committee (listed at https://www.kidney.org/about/board). strategies to increase living kidney donation. Clin J Am Soc
Accepted May 1, 2021, after editorial review by a Deputy Editor. Nephrol. 2017;12(9):1518-1527.
18. Rodrigue JR, Cornell DL, Kaplan B, Howard RJ. A randomized
trial of a home-based educational approach to increase live
References donor kidney transplantation: effects in blacks and whites. Am J
1. Axelrod DA, Schnitzler MA, Xiao H, et al. An economic Kidney Dis. 2008;51(4):663-670.
assessment of contemporary kidney transplant practice. Am J 19. Ismail SY, Luchtenburg AE, Timman R, et al. Home-based family
Transplant. 2018;18(5):1168-1176. intervention increases knowledge, communication and living

330 AJKD Vol 78 | Iss 3 | September 2021


Lentine et al

donation rates: a randomized controlled trial. Am J Transplant. 38. Lentine KL. Pro: Pretransplant weight loss: yes. Nephrol Dial
2014;14(8):1862-1869. Transplant. 2015;30(11):1798-1803.
20. Schweitzer EJ, Yoon S, Hart J, et al. Increased living donor 39. Modanlou KA, Muthyala U, Xiao H, et al. Bariatric surgery
volunteer rates with a formal recipient family education pro- among kidney transplant candidates and recipients: analysis of
gram. Am J Kidney Dis. 1997;29(5):739-745. the United States renal data system and literature review.
21. Grams ME, Sang Y, Levey AS, et al. Kidney-failure risk pro- Transplantation. 2009;87(8):1167-1173.
jection for the living kidney-donor candidate. N Engl J Med. 40. Orandi BJ, Purvis JW, Cannon RM, et al. Bariatric surgery to
2016;374(5):411-421. achieve transplant in end-stage organ disease patients: a sys-
22. Grams ME, Garg AX, Lentine KL. Kidney-failure risk projection tematic review and meta-analysis. Am J Surg. 2020;220(3):
for the living kidney-donor candidate. N Engl J Med. 566-579.
2016;374(21):2094-2095. 41. Kobashigawa J, Dadhania D, Bhorade S, et al. Report from the
23. Lentine KL, Mannon RB. Apolipoprotein L1: role in the evalu- American Society of Transplantation on frailty in solid organ
ation of kidney transplant donors. Curr Opin Nephrol Hyper- transplantation. Am J Transplant. 2019;19(4):984-994.
tens. 2020;29(6):645-655. 42. Cheng XS, Lentine KL, Koraishy FM, Myers J, Tan JC. Impli-
24. Lentine KL, Kasiske BL, Levey AS, et al. KDIGO Clinical cations of frailty for peritransplant outcomes in kidney trans-
Practice Guideline on the evaluation and care of living kidney plant recipients. Curr Transplant Rep. 2019;6(1):16-25.
donors. Transplantation. 2017;101(8S)(suppl 1):S1-S109. 43. Harhay MN, Rao MK, Woodside KJ, et al. An overview of frailty
25. Kasiske BL, Asrani SK, Dew MA, et al. The Living Donor Col- in kidney transplantation: measurement, management and
lective: a scientific registry for living donors. Am J Transplant. future considerations. Nephrol Dial Transplant. 2020;35(7):
2017;17(12):3040-3048. 1099-1112.
26. Kasiske BL, Ahn YS, Conboy M, et al. Outcomes of 44. Cooper M, Formica R, Friedewald J, et al. Report of
living kidney donor candidate evaluations in the Living National Kidney Foundation consensus conference to
Donor Collective pilot registry. Transplant Direct. decrease kidney discards. Clin Transplant. 2019;33(1):
2021;7(5):e689. e13419.
27. Lentine KL, Mandelbrot D. Addressing disparities in living donor 45. Reese PP, Harhay MN, Abt PL, Levine MH, Halpern SD. New
kidney transplantation: a call to action. Clin J Am Soc Nephrol. solutions to reduce discard of kidneys donated for trans-
2018;13(12):1909-1911. plantation. J Am Soc Nephrol. 2016;27(4):973-980.
28. Tushla L, Rudow DL, Milton J, et al. Living-donor 46. Schold JD, Meier-Kriesche HU. Which renal transplant candi-
kidney transplantation: reducing financial barriers to dates should accept marginal kidneys in exchange for a shorter
live kidney donation–recommendations from a consensus waiting time on dialysis? Clin J Am Soc Nephrol. 2006;1(3):
conference. Clin J Am Soc Nephrol. 2015;10(9): 532-538.
1696-1702. 47. Aubert O, Reese PP, Audry B, et al. Disparities in
29. Habbous S, Woo J, Lam NN, et al. The efficiency of evaluating acceptance of deceased donor kidneys between the
candidates for living kidney donation: a scoping review. United States and France and estimated effects of increased
Transplant Direct. 2018;4(10):e394. US acceptance. JAMA Intern Med. 2019;179(10):1365-1374.
30. Habbous S, Barnieh L, Klarenbach S, et al. Evaluating 48. Massie AB, Luo X, Chow EK, Alejo JL, Desai NM, Segev DL.
multiple living kidney donor candidates simultaneously is more Survival benefit of primary deceased donor transplantation with
cost-effective than sequentially. Kidney Int. 2020;98(6):1578- high-KDPI kidneys. Am J Transplant. 2014;14(10):2310-2316.
1588. 49. Husain SA, Chiles MC, Lee S, et al. Characteristics and per-
31. Organ Procurement and Transplantation Network (OPTN). formance of unilateral kidney transplants from deceased do-
Current U.S. waiting list. Accessed March 10, 2021. https:// nors. Clin J Am Soc Nephrol. 2018;13(1):118-127.
optn.transplant.hrsa.gov/data/view-data-reports/national-data/# 50. Lentine KL, Naik AS, Schnitzler MA, et al. Variation in use of
32. Grams ME, Massie AB, Schold JD, Chen BP, Segev DL. procurement biopsies and its implications for discard of
Trends in the inactive kidney transplant waitlist and implications deceased donor kidneys recovered for transplantation. Am J
for candidate survival. Am J Transplant. 2013;13(4):1012- Transplant. 2019;19(8):2241-2251.
1018. 51. Mohan S, Foley K, Chiles MC, et al. The weekend effect alters
33. Huang E, Shye M, Elashoff D, Mehrnia A, Bunnapradist S. the procurement and discard rates of deceased donor kidneys
Incidence of conversion to active waitlist status among in the United States. Kidney Int. 2016;90(1):157-163.
temporarily inactive obese renal transplant candidates. Trans- 52. Schold JD, Buccini LD, Srinivas TR, et al. The association of
plantation. 2014;98(2):177-186. center performance evaluations and kidney transplant volume
34. Ng YH, Pankratz VS, Leyva Y, et al. Does racial disparity in in the United States. Am J Transplant. 2013;13(1):67-75.
kidney transplant waitlisting persist after accounting for social 53. Axelrod DA, Malinoski D, Patel MS, et al. Modeling the eco-
determinants of health? Transplantation. 2020;104(7):1445- nomic benefit of targeted mild hypothermia in deceased donor
1455. kidney transplantation. Clin Transplant. 2019;33(7):e13626.
35. Nishio-Lucar AG, Locke J, Kumar V. Use of patient navigators to 54. Buchanan PM, Schnitzler MA, Axelrod D, Salvalaggio PR,
reduce barriers in living donation and living donor trans- Lentine KL. The clinical and financial burden of early dialysis
plantation. Curr Transpl Rep. 2020;7(2):72-80. after deceased donor kidney transplantation. J Nephrol Ther.
36. Kataria A, Gowda M, Lamphron BP, Jalal K, Venuto RC, 2012;2012(suppl 4).
Gundroo AA. The impact of systematic review of status 7 pa- 55. Schwantes IR, Schnitzler MA, Lentine KL, Balakrishnan R, Reed AI,
tients on the kidney transplant waitlist. BMC Nephrol. Axelrod DA. A simple risk-based reimbursement system for kidney
2019;20(1):174. transplant. Clin Transplant. 2021;35(1):e14068.
37. Lentine KL, Delos Santos R, Axelrod D, Schnitzler MA, 56. Ros RL, Kucirka LM, Govindan P, Sarathy H, Montgomery RA,
Brennan DC, Tuttle-Newhall JE. Obesity and kidney transplant Segev DL. Patient attitudes toward CDC high infectious risk
candidates: how big is too big for transplantation? Am J donor kidney transplantation: inferences from focus groups.
Nephrol. 2012;36(6):575-586. Clin Transplant. 2012;26(2):247-253.

AJKD Vol 78 | Iss 3 | September 2021 331


Lentine et al

57. Organ Procurement and Transplantation Network (OPTN). Align 73. DiRito JR, Hosgood SA, Tietjen GT, Nicholson ML. The future
OPTN Policy with US PHS Guideline 2020. Board Action: of marginal kidney repair in the context of normothermic
December 2020. Accessed April 17, 2021. https://optn. machine perfusion. Am J Transplant. 2018;18(10):
transplant.hrsa.gov/governance/policy-notices/ 2400-2408.
58. Goldberg DS, Abt PL, Blumberg EA, et al. Trial of trans- 74. Fiori S, Remuzzi G, Casiraghi F. Update on mesenchymal
plantation of HCV-infected kidneys into uninfected recipients. stromal cell studies in organ transplant recipients. Curr Opin
N Engl J Med. 2017;376(24):2394-2395. Organ Transplant. 2020;25(1):27-34.
59. Lentine KL, Peipert JD, Alhamad T, et al. Survey of clinician 75. Dharnidharka VR, Schnitzler MA, Chen J, et al. Differential risks
opinions on kidney transplantation from hepatitis C virus posi- for adverse outcomes 3 years after kidney transplantation
tive donors: identifying and overcoming barriers. Kidney360. based on initial immunosuppression regimen: a national study.
2020;1(11):1291-1299. Transpl Int. 2016;29(11):1226-1236.
60. Lentine KL, Singh N, Woodside KJ, et al. COVID-19 test 76. Cheungpasitporn W, Lentine KL, Tan JC, et al. Immunosup-
reporting for deceased donors: emergent policies, logistical pression considerations for older kidney transplant recipients.
challenges and future directions. Clin Transplant. 2021;35(5): Curr Transpl Rep. 2021;8:100-110.
e14280. 77. Lentine KL, Cheungpasitporn W, Xiao H, et al. Immunosuppres-
61. Neidlinger NA, Smith JA, D’Alessandro AM, et al. Organ re- sion regimen use and outcomes in older and younger adult kidney
covery from deceased donors with prior COVID-19: a case transplant recipients: a national registry analysis. Transplantation.
series. Transpl Infect Dis. 2020;23(2):e13503. 2021;105(8):1840-1849.
62. Jan MY, Jawed AT, Barros N, et al. A national survey of practice 78. Oetting WS, Wu B, Schladt DP, et al. Genetic variants asso-
patterns for accepting living kidney donors with prior covid-19. ciated with immunosuppressant pharmacokinetics and adverse
Kidney Int Reports. Published online May 15, 2021;doi:10. effects in the DeKAF Genomics Genome-wide Association
1016/j.ekir.2021.05.003. Studies. Transplantation. 2019;103(6):1131-1139.
63. Bae S, Massie AB, Luo X, Anjum S, Desai NM, Segev DL. 79. Vincenti F, Charpentier B, Vanrenterghem Y, et al. A phase III
Changes in discard rate after the introduction of the Kidney study of belatacept-based immunosuppression regimens
Donor Profile Index (KDPI). Am J Transplant. 2016;16(7):2202- versus cyclosporine in renal transplant recipients (BENEFIT
2207. study). Am J Transplant. 2010;10(3):535-546.
64. Reese PP, Aubert O, Naesens M, et al. Assessment of the utility 80. Busque S, Scandling JD, Lowsky R, et al. Mixed chimerism and
of kidney histology as a basis for discarding organs in the acceptance of kidney transplants after immunosuppressive
United States: a comparison of international transplant prac- drug withdrawal. Sci Transl Med. 2020;12(528).
tices and outcomes. J Am Soc Nephrol. 2021;32(2):397-409. 81. Tantisattamo E, Leventhal JR, Mathew JM, Gallon L. Chimerism
65. Stewart ZA, Shah SA, Formica RN, et al. A call to action: and tolerance: past, present and future strategies to prolong
feasible strategies to reduce the discard of transplantable kid- renal allograft survival. Curr Opin Nephrol Hypertens.
neys in the United States. Clin Transplant. 2020;34(9):e13990. 2021;30(1):63-74.
66. Lentine KL, Kasiske BL, Axelrod D. Procurement biopsies 82. Mannon RB, Morris RE, Abecassis M, et al. Use of biomarkers
in kidney transplantation: More information may not lead to to improve immunosuppressive drug development and out-
better decisions. J Am Soc Nephrol. 2021;32(7):ASN.20210 comes in renal organ transplantation: A meeting report. Am J
30403. Transplant. 2020;20(6):1495-1502.
67. Trial to define the benefits and harms of deceased donor kidney 83. Warady BA, Chadha V. Chronic kidney disease in children:
procurement biopsies. ClinicalTrials.gov identifier: the global perspective. Pediatr Nephrol. 2007;22(12):1999-
NCT03837522. Accessed April 17, 2021. https://clinicaltrials. 2009.
gov/ct2/show/NCT03837522 84. Seikaly MG, Ho PL, Emmett L, Fine RN, Tejani A. Chronic renal
68. Kox J, Moers C, Monbaliu D, et al. The benefits of hypothermic insufficiency in children: the 2001 Annual Report of the
machine preservation and short cold ischemia times in NAPRTCS. Pediatr Nephrol. 2003;18(8):796-804.
deceased donor kidneys. Transplantation. 2018;102(8):1344- 85. North American Pediatric Renal Trials and Collaborative Studies.
1350. NAPRTCS 2014 Annual Transplant Report. North American
69. Gill J, Dong J, Eng M, Landsberg D, Gill JS. Pulsatile perfusion Pediatric Renal Trials and Collaborative Studies; 2014.
reduces the risk of delayed graft function in deceased donor 86. Dharnidharka VR, Fiorina P, Harmon WE. Kidney transplantation
kidney transplants, irrespective of donor type and cold ischemic in children. N Engl J Med. 2014;371(6):549-558.
time. Transplantation. 2014;97(6):668-674. 87. Harshman LA, Johnson RJ, Matheson MB, et al. Academic
70. Pearson R, Asher J, Jackson A, Mark PB, Shumeyko V, achievement in children with chronic kidney disease: a report
Clancy MJ. Viability assessment and utilization of declined from the CKiD cohort. Pediatr Nephrol. 2019;34(4):689-
donor kidneys with rhabdomyolysis using ex vivo normothermic 696.
perfusion without preimplantation biopsy. Am J Transplant. 88. Lande MB, Mendley SR, Matheson MB, et al. Association of
2021;21(3):1317-1321. blood pressure variability and neurocognition in children with
71. Hosgood SA, Saeb-Parsy K, Wilson C, Callaghan C, Collett D, chronic kidney disease. Pediatr Nephrol. 2016;31(11):2137-
Nicholson ML. Protocol of a randomised controlled, open-label 2144.
trial of ex vivo normothermic perfusion versus static cold stor- 89. Solomon MA, van der Plas E, Langbehn KE, et al. Early pedi-
age in donation after circulatory death renal transplantation. atric chronic kidney disease is associated with brain volumetric
BMJ Open. 2017;7(1):e012237. gray matter abnormalities. Pediatr Res. 2021;89(3):526-532.
72. Meister FA, Czigany Z, Bednarsch J, et al. Hypothermic 90. Foster BJ, Dahhou M, Zhang X, Platt RW, Samuel SM,
oxygenated machine perfusion of extended criteria kidney Hanley JA. Association between age and graft failure rates in
allografts from brain dead donors: protocol for a prospec- young kidney transplant recipients. Transplantation.
tive pilot study. JMIR Res Protoc. 2019;8(10):e14622. 2011;92(11):1237-1243.

332 AJKD Vol 78 | Iss 3 | September 2021

You might also like