Cell-Free DNA Diagnostics in Transplantation Utilizing Next Generation Sequencing

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Human Immunology 82 (2021) 850–858

Contents lists available at ScienceDirect

www.ashi-hla.org

journal homepage: www.elsevier.com/locate/humimm

Review

Cell-free DNA diagnostics in transplantation utilizing next generation


sequencing
Annette M. Jackson a,⇑, Carly Amato-Menker b, Maria Bettinotti c
a
Duke University, Department of Surgery, DUMC Box 2645, Durham, NC 27710, USA
b
West Virginia University, Microbiology, Immunology, and Cell Biology, Morgantown, WV, USA
c
Johns Hopkins University, Department of Pathology, 2041 E. Monument Street, Baltimore, MD 21205, USA

a r t i c l e i n f o a b s t r a c t

Article history: The use of Next Generation Sequencing (NGS) to interrogate cell-free DNA (cfDNA) as a transplant diag-
Received 14 April 2021 nostic provides a crucial step in improving the accuracy of post-transplant monitoring of allograft health.
Revised 29 June 2021 cfDNA interrogation provides a powerful, yet minimally invasive, biomarker for disease and tissue injury.
Accepted 12 July 2021
cfDNA can be isolated from a variety of body fluids and analyzed using bioinformatics to unlock its ori-
Available online 30 September 2021
gins. Furthermore, cfDNA characteristics can reveal the mechanisms and conditions under which it was
generated and released. In transplantation, donor-derived cfDNA monitoring provides a tool for identify-
Keywords:
ing active allograft injury at the time of transplant, infection, and rejection. Multiple detection and inter-
Transplantation
Rejection
rogation methods for cfDNA detection are now being evaluated for clinical validity and hold the promise
Cell free DNA to provide minimally invasive, quantitative, and reproducible measures of allograft injury across organ
DNA sequencing types.
Biomarker Ó 2021 American Society for Histocompatibility and Immunogenetics. Published by Elsevier Inc. All rights
reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 850
2. Characteristics of circulating cfDNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 851
3. Collection and processing of Cir-cfDNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 852
4. Detection of donor derived Cir-cfDNA as a transplant rejection diagnostic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 853
5. Deciphering the cellular origins of Cir-cfDNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 854
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 856
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 856
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 856

Abbreviations: ABMR, antibody mediated rejection; ACR, acute cellular rejec-


tion; AR, acute rejection; AUC, area under the curve; bp, nucleotide base pairs; 1. Introduction
cfDNA, cell-free DNA; dd-Cir-cfDNA, donor derived circulating cell-free DNA; CV,
coefficient of variance; DSA, donor-specific HLA antibody; HHV4-8, human Cell-free DNA (cfDNA) are fragments of degraded DNA released
herpesviruses types 4-8; NPV, Negative predictive values; NGS, Next Generation primarily from cells undergoing apoptosis. Due to its rapid turnover,
DNA Sequencing; PCR, polymerase chain reaction; PPV, positive predictive values;
cfDNA circulating in blood (Cir-cfDNA) provides a window into the
STR, short tandem repeat; SNP, single nucleotide polymorphism; TCMR, T cell
mediated rejection. physiology and pathology of cells or tissues in real time. Cir-cfDNA
⇑ Corresponding author. analysis has been adopted as a valuable clinical laboratory
E-mail address: annette.m.jackson@duke.edu (A.M. Jackson).

https://doi.org/10.1016/j.humimm.2021.07.006
0198-8859/Ó 2021 American Society for Histocompatibility and Immunogenetics. Published by Elsevier Inc. All rights reserved.
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

Fig. 1. Cell-free DNA nomenclature and diagnostic utility. Cell-free DNA can be isolated from a variety of human body fluids and utilized for different diagnostic purposes.
This diversity led the 10th International Circulating Nucleic Acids in Plasma and Serum consortium to propose common nomenclature. This figure is being used with author’s
permission [7].

diagnostic and monitoring tool across diverse disciplines such as appreciation for the need for more sensitive methods to diagnose
oncology, prenatal screening, infectious disease, and transplantation. rejection at its earliest stages prior to clinical allograft dysfunction.
The common thread is the need for precision medicine and non- Histopathology remains the gold standard for rejection diagnoses
invasive sampling to allow for accurate and frequent monitoring. and provides important phenotypic and cumulative injury per-
cfDNA can be isolated from a variety of human body fluids such spectives. However, currently the detection of early subclinical
as blood, urine, saliva, lymph, breastmilk, bile, spinal and amniotic rejection requires monitoring through serial surveillance biopsies,
fluid and serves as a non-invasive biomarker for disease, infection, the majority of which reveal no evidence of rejection. The develop-
and tissue injury [1–5]. Moreover, detectable cfDNA may originate ment of non-invasive diagnostic tools such dd-Cir-cfDNA assays
from self or from non-self sources such as fetal tissue, microbial may provide a complementary tool for detecting early, subclinical
and viral DNA, or transplanted stem cells and solid organ allografts. rejection and also provide information regarding rejection type
All individuals harbor baseline levels of cfDNA that derive primar- (antibody mediated rejection (ABMR) versus T cell mediated rejec-
ily from ongoing cell turn-over, however the quantity and compo- tion (TCMR)) to guide appropriate interventions.
sition of cfDNA can vary. Daily activities such as exercise have been This review will explore our current understanding of Cir-
shown to impact circulating plasma proteins levels to include Cir- cfDNA characteristics and current practices for isolation and inter-
cfDNA [6]. Importantly, interrogation of cfDNA characteristics rogation using next generation sequencing (NGS) platforms. We
holds the potential to reveal the source and the mechanisms and examined the current literature for clinical studies utilizing Cir-
conditions under which cfDNA is generated and released [7]. This cfDNA in the setting of transplantation to highlight the current
diversity in cfDNA profiles has led members of the 10th Interna- strengths and limitations of this methodology. Finally, looking to
tional Circulating Nucleic Acids in Plasma and Serum consortium the future, we discuss how Cir-cfDNA assays may evolve to reveal
to recommend a unified cfDNA terminology and nomenclature to greater details surrounding allograft injury and aid in the complex
clarify the source of cfDNA in publications and clinical reporting differential diagnosis of allograft rejection.
Fig. 1 [8]. Through bioinformatic analysis of cfDNA, laboratories
can unlock the cell origins and the state of tissue health to advance 2. Characteristics of circulating cfDNA
non-invasive, personalized medicine.
In the field of solid organ transplantation, detection of donor The concentration of Cir-cfDNA in healthy individuals is low
derived circulating cfDNA (dd-Cir-cfDNA) is being explored as a and a recent study of 84 healthy subjects revealed a range of 1–
non-invasive measure of allograft injury Fig. 2. There is a growing 10 ng/ml in plasma, with an average of 6 ng/ml reflecting a geno-
851
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

Fig. 2. Detection of donor circulating cell-free DNA in transplant diagnostics. Detection of donor derived circulating cfDNA (dd-Cir-cfDNA) is being explored as a non-invasive,
diagnostic measure of allograft injury. Informative single nucleotide polymorphism (SNP) differences between recipient and donor genotypes can be leveraged to allow the
detection and quantification of dd-Cir-cfDNA. In plasma, the majority of Cir-cfDNA molecules derive from the recipient (blue) and a minor fraction derive from donor cells
(black). Detection of increased levels of dd-Cir-cfDNA above baseline indicates allograft injury. (For interpretation of the references to colour in this figure legend, the reader is
referred to the web version of this article.)

mic DNA equivalent of 1000 cells [9]. The half-life of Cir-cfDNA in derived from the recipient (165 bp) [15]. Similarly, fetal derived
plasma is short and estimated to be between 10 min and 2.5 h [18]. Cir-cfDNA molecules tend to be smaller (143 bp) than prominent
Removal is facilitated primarily by the liver, but nuclease activity, 166 bp maternal fragments [16] and tumor associated Cir-cfDNA
degradation by splenic macrophages, and renal excretion also play molecules smaller (134–144 bp) than Cir-cfDNA from healthy con-
a role [7]. Detection of elevated levels of Cir-cfDNA therefore trols (167 bp) [10,12]. Given that hematopoietic derived blood cells
reflects an active and excessive release of DNA caused by extensive contribute the majority of the plasma Cir-cfDNA, the smaller Cir-
cell death, inefficient removal of the dead cells, or a combination of cfDNA fragments observed from transplants, tumors, and fetal tis-
both. sue likely reflect tissue-specific differences in nucleosome wrap-
Cell apoptosis is considered the primary source of Cir-cfDNA ping which may influence Cir-cfDNA fragment lengths deriving
within plasma or serum and this is consistent with paired-end from different tissues [12,14,15].
sequencing data showing the dominant DNA species being
nucleosome-size DNA fragments comprising 160–180 base pairs 3. Collection and processing of Cir-cfDNA
(bp) [10,11]. Nucleosome packaging of DNA involves 147 bp
wrapped around a histone octamer with an additional linker The sensitivity needed for Cir-cfDNA interrogation in many clin-
DNA of approximately 20–90 bp [12]. This nucleosomal structure ical settings requires considerable attention to technical details
stabilizes DNA in the circulation and provides protection from surrounding sampling and processing procedures. One of the pos-
enzymatic degradation. Larger Cir-cfDNA fragments have been sible sources of interference is shedding of genomic DNA from
reported and are speculated to come from necrotic tissue, while blood cells after sample collection. Plasma is the preferred Cir-
mitochondrial DNA is also found in the Cir-cfDNA fraction and cfDNA source over serum due to this contamination of genomic
the size distribution of this non-histone-bound form is consistently DNA from leukocytes during the clotting process. Raymond et al.
shorter [10]. examined the impact of time between blood collection and plasma
The mean concentration of Cir-cfDNA has been reported to be processing using replicate samples spiked with a Cir-cfDNA stan-
higher in healthy men when compared to healthy women, which dard and processed over 7 days. Blood was collected using stan-
may reflect differences in body mass [13]. Importantly, patients dard ethylenediaminetetraacetic acid (EDTA) anticoagulant. This
with autoimmune disease, cancer, recent transplants, or pregnancy study showed that longer processing times did not significantly
exhibit elevated Cir-cfDNA compared to healthy individuals interfere with the detection of the targeted Cir-cfDNA. A second
reflecting their altered medical state and/or tissue injury [10,14]. study of 106 normal controls using whole blood stored in EDTA
Interestingly, paired-end sequence analyses of Cir-cfDNA mole- tubes at +4 °C showed no change in Cir-cfDNA concentration after
cules deriving from liver allografts have been found to be slightly one day of storage, validating the potential for overnight ship-
smaller (110–140 bp) than predominant Cir-cfDNA molecules ments of samples in this preservative [17]. Nevertheless, in the

852
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

clinical setting, blood collection tubes that contain stabilizers to 4. Detection of donor derived Cir-cfDNA as a transplant
prevent cell lysis remain a preferred choice for blood collection. rejection diagnostic
The extraction method used to isolate Cir-cfDNA from plasma
can also impact the yield and fragment size bias [18–21]. Compar- The advancement of molecular diagnostic techniques and bioin-
isons have shown that extraction procedures specific for Cir-cfDNA formatics has led to an evolution of methods to detect and charac-
isolation out-perform standard genomic DNA extraction proce- terize Cir-cfDNA within the transplant setting [3,24–27]. Early
dures in providing a better representation of smaller DNA frag- studies utilized multiplex, quantitative polymerase chain reaction
ments from larger plasma volumes (1–4 ml). Details such as the (PCR), followed by the development of digital drop microfluidics
number and speed of centrifugation steps to isolate Cir-cfDNA PCR and NGS methods. Development of sensitive and efficient
from plasma has been found to be important. An initial low g- methods for detecting donor derived Cir-cfDNA (dd-Cir-cfDNA)
force centrifugation step is performed to separate plasma from released from transplanted allografts promises to provide a mini-
cells and a second, higher g-force centrifugation removes all mally invasive, quantitative, and reproducible measure of allograft
remaining cellular debris. The speed of the second centrifugation injury across organ types.
step may influence recovery of different nucleic acid types. Higher Transplanted allografts are injured at multiple points during the
centrifugation speeds (e.g., 16,000g) aid in removal of intact transplant process and it is the cumulative injury that likely
genomic DNA from ruptured blood cells, but could also remove impacts long-term survival potential [28–33]. Peri-operatively
large extracellular vesicles that may contain RNA and cfDNA. Page allografts sustain injury during procurement, cold storage, and at
et al. examined different kits and different speeds for the second time of reperfusion. Acute rejection is also common, particularly
centrifugation step (1,000g, 2,000g, or 10,000g for 10 min at within the first year post-transplant. Infection is likely an underap-
4 °C) and found yields of cfDNA to be equivalent, as assessed by preciated form of allograft injury except in cases of severe infection
locus-specific TaqMan quantitation of GAPDH, while miRNA yield coinciding with allograft dysfunction. A multidisciplinary inves-
was lower after centrifugation at 10,000g [19]. For clinical diag- tigative group at Stanford University established a ‘‘shotgun” or
nostics, the extraction procedure, collection tube, and minimal random DNA NGS sequencing approach to monitor dd-Cir-cfDNA
plasma starting volumes should be optimized for the specific in heart and lung transplant recipients by quantifying tens-of-
detection method, analyte, and within specific study populations. thousands of reads containing informative single nucleotide poly-
DNA quantification and input for library construction is also an morphism (SNPs) differentiating donor from recipient [34]. The
important step to ensure the sensitivity in detecting tumor, fetal- estimated informative SNP variations between individuals is 1.6
derived, or allograft derived Cir-cfDNA. Alborelli et al. performed million, using sites where the recipient is homozygous and the
Cir-cfDNA analysis on 114 healthy normal controls to determine donor possesses a different nucleotide on one or both or alleles
feasibility of cancer screening in populations expected to have [35]. Each sequencing read containing an informative SNP is char-
low Cir-cfDNA levels [22]. cfDNA was extracted from between acterized as recipient, donor, or PCR artifact and from these data
0.4 and 2.0 ml of plasma using a magnetic beads-based cfDNA iso- the percentage of dd-Cir-cfDNA to recipient Cir-cfDNA is calcu-
lation kit yielding 1.7 to 30.8 ng/ml of cfDNA. Quantification was lated. Therefore, for these studies both the recipient and donor
performed using a Qubit Fluorometer followed by genotypes are needed to distinguish donor-specific sequencing
electrophoresis-based size selection (Agilent High Sensitivity reads. Analyses of longitudinal post-transplant plasma samples
D1000 ScreenTape System). Adequate Cir-cfDNA quantity and obtained from heart and lung recipients showed high levels of
quality to proceed to NGS library preparation was achieved for dd-Cir-cfDNA on the first day post-transplant, followed by an
only 55 out of 114 normal controls. NGS libraries were prepared exponential decay to a low, steady state baseline level [36,37].
using a broad range of cfDNA input (2.5–105.5 ng), with acceptable Organ-specific differences were observed in the amount of dd-
results in the lower concentrations, which were far below the min- Cir-cfDNA detected immediately post-transplant and in the decay
imal manufacturers’ recommended amount of 10 ng. kinetics. Consistent with organ mass, dd-Cir-cfDNA was higher in
Double-strand library preparation is standard for Cir-cfDNA bilateral compared to single-lung transplants and higher in lung
analysis because of efficiency, cost, and the optimization of meth- transplants compared to heart transplants. The heart transplant
ods to avoid adapter ligation bias. However, for some applications data showed single decay kinetics (single exponential), while the
such as the paleontological field working with ancient DNA, the lung transplant data showed a two-step decay model during the
value of single-strand library preparation to recover damaged first weeks post-transplant.
and short double-strand DNA fragments, normally lost in double- Studies have also demonstrated elevations in dd-Cir-cfDNA
strand library preparation, has proven important [23]. Bennet levels above baseline occurring at time of infection [36–39]. Ran-
et al. optimized a single-strand library method using a 50 - dom shotgun sequencing allows data interrogation for pathogen-
phosphorylated and biotinylated adapter oligonucleotide and a derived sequences and extends the scope of Cir-cfDNA monitoring
single-strand DNA ligase to capture single-strand DNA molecules to include infectious disease detection. For lung recipients
onto beads without prior end repair. A primer generated comple- elevated dd-Cir-cfDNA correlated with clinically diagnosed cyto-
mentary strands, followed by a second adaptor addition via megalovirus infection in serum or lung lavage fluid with an area
blunt-end ligation. The molecules are finally heated to release under the curve (AUC) = 0.91 [37]. High frequency clinical and
the finished single strand, which is used in the adaptor sequence subclinical infections were also detected involving polyomavirus,
amplification reaction. Cir-cfDNA molecules that are damaged, human herpesviruses (HHV4-8) and adenovirus as well as bacte-
for example with single-strand breaks on one or both strands, rial and fungal infections, some of which are not routinely tested
are retained within this single-strand library. In a study utilizing for post-transplant.
Cir-cfDNA isolated from cancer patients, Sachez et al. compared Detection of acute rejection in the early subclinical stages, prior
double-strand and single-strand library preparations and found to injury sufficient to yield dysfunction, and early identification of
nearly half of the total Cir-cfDNA fragments were<130 nucleotides, chronic rejection lesions would provide opportunities for interven-
ranging from 30 to 130, and were not detectable using the double- tion in the hopes of improving long-term allograft survival. The
strand library preparations [12]. Given that Cir-cfDNA fragmenta- current ‘‘gold standard” diagnostic for acute and chronic rejection
tion may be variable depending on the disease state, optimal remains the histological grading of biopsy tissue. However,
recovery of smaller Cir-cfDNA molecules may be important to histopathology has inherent limitations relating to sensitivity,
ensure sensitivity and specificity in clinical diagnostics. inter-observatory variability, and dependence on qualitative not
853
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

quantitative measures. Therefore, determining clinical validity of cfDNA across diverse study cohorts. The prevalence of rejection
more sensitive and potentially more accurate rejection diagnostics in each cohort can impact the diagnostic acumen of the assay;
will require rigorous validation studies using long-term allograft increased rejection prevalence in higher risk cohorts will increase
function and survival as clinical endpoints. Agbor-Enoh et al. inde- the PPV but decrease the NPV. Similarly, lower rejection prevalence
pendently validated and applied the Stanford Shotgun NGS method decreases the PPV while increasing the NPV [58]. Given the low
using a 1% threshold for dd-Cir-cfDNA and longitudinal samples PPV and current limitations in our understanding of how to inter-
from lung and heart recipients [32,38,40,41]. These studies added pret and whom and when to monitor for dd-Cir-cfDNA, this assay
to previous reports showing the utility of dd-Cir-cfDNA to detect should not be considered a stand-alone diagnostic. Rather the high
injury at time-points preceding clinical histopathological diag- NPV signifies utility as an integrated screening test to be inter-
noses for rejection and before clinical dysfunction [36,37]. The preted with other clinical parameters, immunologic risk categories,
Stanford collaborative group have further developed a bioinfor- and companion diagnostics from infectious disease and histocom-
matics algorithm to identify minority donor sequencing reads patibility laboratories.
and thereby eliminate the need for full recipient and donor Newer studies are evaluating dd-Cir-cfDNA measurements calcu-
genotype information [42]. Nevertheless, removal of the actual lated as absolute concentrations (cp/mL) versus the conventional per-
donor and recipient genotype information from the bioinformat- cent fraction (%) of recipient Cir-cfDNA. Zhao et al. measured dd-Cir-
ics increases the need to guard against false positive results cfDNA in 49 pediatric liver recipients and observed similar correla-
stemming from blood products or cases of multi-organ or tions with biopsy proven rejection (n = 11) whether using absolute
re-transplantation. concentration (AUC = 0.841) or percent fraction (AUC = 0.878) [39].
The current commercial assays including those in clinical trials In contrast, Oellerich et al. performed a longitudinal analysis of 189
use multiplex PCR coupled with NGS to specifically target hun- kidney recipients and showed that the absolute concentration of
dreds to thousands of informative SNPs to detect dd-Cir-cfDNA. dd-Cir-cfDNA (AUC = 0.83), when detected using digital droplet
In these assays, the predominant, informative SNPs are assigned PCR, outperformed the percent fraction (AUC = 0.73) in discriminating
to the recipient and the minority fraction are arbitrarily assigned rejection (n = 15) from stable allograft function [59]. One potential
as dd-Cir-cfDNA using algorithms that do not require recipient or reason for this latter finding and a potential advantage for calculating
donor genotypes. Grskovic et al. reported on the precision of dd- absolute concentrations is the finding that exercise can increase Cir-
Cir-cfDNA detection using a NGS targeted sequencing of 266 infor- cfDNA derived from hematopoietic cells in the blood, resulting in
mative SNPs [43]. Assay validation utilized 1,117 samples from the higher recipient cfDNA levels and a potential underestimation of
National Institute for Standards and Technology Genome in a Bot- dd-Cir-cfDNA levels when using a percent fraction calculation [20].
tle human reference genome and determined a limit of detection of Two studies, in kidney and lung, show the power of combining
0.16% and limit of quantification of 0.20% dd-Cir-cfDNA. The linear dd-Cir-cfDNA and HLA antibody detection methods. Jordan et al.
quantifiable range was 0.2%  16% and run-to-run coefficient of investigated the utility of dd-Cir-cfDNA to aid in ABMR diagnoses
variance (CV) was 6.8%. Bromberg et al. reported further using this when combined with donor-specific HLA antibody (DSA) testing
same assay to examine dd-Cir-cfDNA biological variation within [48]. Eighty-seven kidney recipients were examined using 90 blood
380 longitudinal blood samples from 93 stable transplant recipi- samples with paired DSA and biopsies performed due to clinical
ents [44]. The median dd-Cir-cfDNA was 0.21% (interquartile range suspicion. The PPV of dd-Cir-cfDNA (1% positive threshold) to
0.12%–0.39%) and 96% of the samples exhibited values below 1.0%. detect active ABMR in 16 DSA positive patients was 81% and the
Of note, these studies were performed using a centralized labora- NPV was 83%. In comparison, the ABMR PPV for DSA + alone was
tory and further interlaboratory studies will be needed if siimilar 48%. This study highlights the use of multiple biomarkers to
assays are released to transplant laboratories in a kit form. increase diagnostic accuracy. dd-Cir-cfDNA is a sensitive short-
Retrospective and prospective clinical studies have been per- lived biomarker of allograft injury, but is not specific for rejection.
formed in kidney, heart, lung, and liver transplant cohorts and DSA holds a much longer half-life (~30 days) in the blood and sig-
are summarized in Table 1 [36,37,39,41,45–57]. Rejection preva- nifies an active alloimmune response, but at low levels DSA may
lence and rejection type varied between studies and different % not correlate with significant tissue injury. Agbor-Enoh et al. also
dd-Cir-cfDNA positive thresholds were used depending on the examined the relationship between DSA and dd-Cir-cfDNA in 34
study endpoint. In spite of these variations, some common patterns lung recipients with active ABMR. All patients had detectable
have emerged. In general, higher dd-Cir-cfDNA thresholds were DSA at time of ABMR diagnosis exhibiting a wide MFI range
sufficient to detect active ABMR whereas lower thresholds were (1896–24,278). Using mixed linear modeling, a significant correla-
needed to correlate with clinical and borderline TCMR. Negative tion was seen between DSA MFI and dd-Cir-cfDNA levels [32].
predictive values (NPV) were high and ranged from 83 to 100% Therefore, utilizing dd-Cir-cfDNA in combination with DSA detec-
across studies and organ types; however, the positive predictive tion may enhance the detection of clinical and subclinical ABMR.
values (PPV) were lower in all cases and varied widely between Furthermore, enhancements in dd-Cir-cfDNA specificity, as
studies Table 1. described in the next section, may increase diagnostic potential
NPV and PPV describe the clinical validity of dd-Cir-cfDNA diag- for subclinical injury that occurs in the absence of clinical dysfunc-
nostic tests, using a set % threshold for positivity and expected tion or histopathological evidence of rejection.
rejection prevalence Fig. 3. NPV represents the specificity of a
screening test to detect all true negatives while PPV estimates
the sensitivity to correctly identify all recipients who have allograft 5. Deciphering the cellular origins of Cir-cfDNA
injury (true positives) [58]. Interpreting these values in the setting
of clinical transplantation requires full recognition of the current Future improvements for Cir-cfDNA diagnostics in transplanta-
limitations to our gold standard (histopathology) for rejection tion will include the ability to trace this biomarker back to the
diagnoses and our current limitations to detect all sources of allo- exact cell types that have been injured and released DNA. This
graft injury (e.g. subclinical rejection, infection and disease recur- advancement would broaden the utility of Cir-cfDNA as a mulit-
rence). The lower PPVs in all of these clinical studies may reflect purpose diagnostic in transplantation, beyond detecting rejection
methodology problems with the assays or misconceptions sur- alone. This capability would provide considerable clarity surround-
rounding the full range of insults that an allograft incurs, making ing the type and mechanism of allograft injury such as antibody
it difficult to accurately assess the clinical validity of dd-Cir- mediated injury of peritubular capillaries versus podocyte injury
854
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

Table 1

Organ type Cohort Rejection prevalence Endpoint Threshold & AUC PPV NPV
Kidney
Bloom, RD et al., J Am Soc N = 102 Biopsy-paired samples = 107 No rejection Threshold 1% AR = 61% =84%
Nephrol, 2017 AR = 27 cases Vs AR AR AUC = 0.74 ABMR = 44% =96%
ABMR = 16; TCMR = 11 Vs ABMR ABMR AUC = 0.87
TCMR = ND
Jordan SC, et al., Transplantation N = 87 Biopsy-paired sample & DSA = 90 DSA+ ABMR Threshold 1% ABMR = 81% =83%
Direct 2018 DSA+ ABMR = 16 cases
Sigdel TK, et al., J Clin Med, 2019 N = 193 Biopsy-paired samples = 217 No rejection Threshold 1%
AR = 38 cases Vs AR AR AUC = 0.87 AR = 51.9% = 95.1%
Borderline AR = 72 cases Borderline AR = No
correlation
Huang E, et al., Am J Transplant, N = 63 Cross sectional No rejection Threshold = .74% ABMR = 68.6% = 100%
2019 analysis = suspicious for rejection Vs TCMR TCMR AUC = 0.42
AR+ = 34 cases Vs ABMR ABMR AUC = 0.82
DSA+ = 27 cases
Gielis EM, et al., Nephrol Dial N = 107 Biopsy-paired samples = 65 No rejection Threshold = 0.88% AR = 8.7% = 97.8%
Transplant, 2020 AR = 13 cases Vs AR AR = AUC 0.64
Stites E, et al., Am J Transplant. N = 79 Cross sectional analysis= eGFR decline Threshold 0.5% NA NA
2020 TCMR 1A & Borderline = 79 cases low cf-DNA <0.5% Multivariate analysis
Vs 6 month -8.5% eGFR decline
High cfDNA >0.5% p = .0040
Puliyanda DP, et al., Ped N = 67 ped Cross sectional analysis = 48 No rejection Threshold 1% NA NA
Transplantation, 2020 cases Vs AR AR AUC = 0.996
suspicious for rejection
OR biopsies w/ dd-cfDNA >1.0%
Zhang H, et al. FrontImmunol N = 37 18=ABMR+, DSA+ No rejection Threshold 1% ABMR = 76.2% = 87.5%
2020 Vs ABMR ABMR AUC = 0.90
Shen J et al. Clin Tranplants, 2020 N = 28 Cross sectional analysis Treatment assoc positive correlation @ NA NA
ABMR = 5 Ddd-cfDNA% respective timepoints
TCMR (1A or1B) = 12 Vs q = 2.570, P = .022
TCMR (2A or 2B) = 11 DeGFR (1,3,6 mo) q = 3.210, P = .027
q = 2.860, P = .019
Heart
De Vlaminck I, et al., Sci Transl N = 21 ped 356 samples paired with No rejection Threshold = .25% NA NA
Med 2014 N = 44 endomyocardial biopsies Vs ACR < 2R/3A Mild AR AUC = 0.63
adult AR = 24 cases Vs ABMR 1 or Mod/Severe AR AUC = 0.83
ACR 2R/3A
Khush KK, et al., Am J Transplant Multi 841 samples paired with No rejection Threshold = .20% ABMR 1 or
2019 Cohorts endomyocardial biopsies Vs ABMR 1 or ACR AUC = 0.64 ACR 2R = 97.1%.
N = 443 >55 days posttransplant 2R AUC = NA = 8.9% = 97.9%
N = 33 ACR = 17 & ABMR = 18 No rejection ABMR 1
High risk 110 plasma samples Vs ABMR 1 = 20.2%
>14 days post-transplant
ABMR = 11
North PE, et al., PLoS One 2020 N = 76 158 samples paired with No rejection Threshold = 0.32% ACR = 43.90% =94.87%
Ped & endomyocardial biopsies Vs ACR 2R AUC = 0.842
Adult 8 days post-transplant
Richmond ME, et al., J Heart Lung N = 241 624 samples paired with No rejection Threshold = 0.16% ACR 1R or
Transplant 2020 Ped & endomyocardial biopsies Vs ACR 1R or AUC = 0.78 ABMR 1 = 85.7%
Adult >7 days post-transplant ABMR 1 = 65.2%
ACR 1 = 15 cases
AMR 1 = 20 cases

Agbor-Enoh S, et al., Circulation N = 171 1,392 endomyocardial biopsies No rejection Threshold = .25% ABMR 1
2021 1,834 plasma samples Vs ABMR 1 or AUC = 0.92 ACR = 99%
ACR 2 = 38 cases ACR 2R 2R = 19.6%
AMR 1 = 32 cases
Mixed AMR/ACR = 3 cases
Lung
De Vlaminck I, et al., Proc Natl N = 51 Biopsy-paired samples = 113 No rejection Threshold 1% NA NA
Acad Sci 2015 >60 days post-transplant Vs severe ACR A3 ACR A3 AUC = 0.90
Vs mild-severe ACR ACR A1 AUC = 0.70
A1
Sayah D, et al., Transplantation N = 69 ACR = 13 No rejection Threshold = 0.87% ACR = 34.1% = 85.5%
Direct 2020 Vs ACR A1 ACR AUC = 0.717
Levine DJ, et al., Biomarker N = 48 CLAD, N = 10 No rejection Threshold = 0.51% NA NA
Insights, 2020 ABMR, N = 9 Vs AR AUC = 0.98
ACR, N = 2
Liver
Zhao D, et al., Scientific Reports N = 49 Biopsy-paired samples = 49 No rejection Threshold = 28.7% AR = 80% = 92.3%
2021 Ped AR=11 Vs AR AUC = 0.878 AR = 56.2% = 93.9%
Threshold = 2076 cp/mL
AUC = 0.841

855
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

Fig. 3. Determining analytical and clinical validity of cell-free-DNA diagnostics. Sensitivity measures the proportion of positive tests that correctly identify rejection (True
Positive) while Specificity measures the proportion of negative tests that correctly identify no rejection (True Negative). Negative and Positive Predictive Values (NPV and
PPV) describe the performance of a diagnostic test using a set threshold for positivity and expected rejection prevalence. NPV represents the specificity of a screening test to
detect all true negatives while PPV estimates the sensitivity to correctly identify all recipients who have allograft rejection (true positives).

correlating with recurrent glomerular disease. Cir-cfDNA monitor- 6. Conclusion


ing may also allow early detection of malignancies in transplant
patients. This could advance the diagnosis of graft-versus-host dis- The addition of dd-Cir-cfDNA detection by NGS as a noninvasive
ease following hematopoietic stem cell transplantation or multi- transplant diagnostic provides a significant opportunity to improve
visceral transplants by detecting elevated recipient tissue injury the detection of rejection, infection, and recurrent disease. Under-
in the gut and skin. Furthermore, Cir-cfDNA diagnostics could be standing the characteristics of Cir-cfDNA impacted by isolation and
extended to the pre-transplant setting for monitoring the progres- processing is crucial to achieve accurate and sensitive results. Cur-
sion of end-stage organ failure or recovery following living organ rent commercial assays that quantitate major (recipient) and
donation. minor (donor) Cir-cfDNA fractions are available, yet further studies
Current clinical diagnostics distinguish Cir-cfDNA origins (self are needed to clinically validate specific endpoint thresholds and
versus non-self) based on the genetic differences (SNPs). This is determine optimal use with companion diagnostics provided by
true for prenatal screening (fetal versus maternal origins), mutated infectious disease and histocompatibility laboratories. Future
tumors versus heathy tissues, and transplant rejection (donor ver- advances in deciphering the exact origins of Cir-cfDNA will further
sus recipient). However, Cir-cfDNA that is derived from different improve its utility and accuracy as a diagnostic tool. Importantly,
cell types often exhibit unique DNA methylation patterns, histone dd-Cir-cfDNA assays hold the potential for non-invasive monitor-
modifications, and nucleosome positioning reflecting cell-specific ing allograft health, providing highly sensitive injury surveillance
gene expression patterns [60–62]. The lack of genetic differences in real time, and facilitating early detection with the possibility
(SNPs) within self-DNA can be overcome by interrogating the of intervention prior to clinical dysfunction. Integrated with clini-
cells-specific epigenetic marks reflecting silent genes (DNA methy- cal parameters, histocompatibility and infectious disease diagnos-
lation) or active genes (histone modifications and nucleosome tics, and allograft function tests, the measurement of dd-Cir-cfDNA
positioning). Cells of the same type will share nucleosome posi- increases our arsenal of tools to improve transplant outcomes.
tioning within specific genes; these nucleosome-protected DNA
sequences will undergo less protease digestion in the blood and Declaration of Competing Interest
will be enriched in the Cir-cfDNA fraction if injury is localized to
one tissue region or cell type. Deep sequencing of Cir-cfDNA can The authors declare that they have no known competing finan-
generate maps of in vivo nucleosome occupancy reflecting the cial interests or personal relationships that could have appeared
cell-specific nuclear architecture and gene expression patterns. to influence the work reported in this paper.
These nucleosome footprints can then be used to infer the cell
types or tissue types contributing to Cir-cfDNA in pathological References
states such as transplant allograft rejection. This type of specificity
and increased sensitivity could aid in the diagnosis of borderline [1] G.C. Di Renzo, J.L. Bartha, C.M. Bilardo, Expanding the indications for cell-free
acute rejection, ABMR versus TCMR versus mixed rejection, and DNA in the maternal circulation: clinical considerations and implications, Am.
J. Obstet. Gynecol. 220 (6) (2019) 537–542.
potentially differentiate rejection from other forms of injury such [2] T.A. Blauwkamp, S. Thair, M.J. Rosen, L. Blair, M.S. Lindner, I.D. Vilfan, T. Kawli,
as recurrent disease. F.C. Christians, S. Venkatasubrahmanyam, G.D. Wall, A. Cheung, Z.N. Rogers, G.

856
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

Meshulam-Simon, L. Huijse, S. Balakrishnan, J.V. Quinn, D. Hollemon, D.K. cell-free DNA for assessment of allograft rejection and injury status, J. Clin.
Hong, M.L. Vaughn, M. Kertesz, S. Bercovici, J.C. Wilber, S. Yang, Analytical and Med. 9 (5) (2020) 1480, https://doi.org/10.3390/jcm9051480.
clinical validation of a microbial cell-free DNA sequencing test for infectious [27] F. Dengu, Next-generation sequencing methods to detect donor-derived cell-
disease, Nat. Microbiol. 4 (4) (2019) 663–674. free DNA after transplantation, Transplant. Rev. (Orlando) 34 (3) (2020)
[3] S.R. Knight, A. Thorne, M.L. Lo Faro, Donor-specific Cell-free DNA as a 100542, https://doi.org/10.1016/j.trre.2020.100542.
biomarker in solid organ transplantation. A systematic review, [28] V. Ronca, G. Wootton, C. Milani, O. Cain, The immunological basis of liver
Transplantation 103 (2) (2019) 273–283. allograft rejection, Front. Immunol. 11 (2020) 2155.
[4] V. Kubaczkova, D. Vrabel, L. Sedlarikova, L. Besse, S. Sevcikova, Cell-free DNA - [29] E. Van Loon, J. Bernards, A.H. Van Craenenbroeck, M. Naesens, The causes of
Minimally invasive marker of hematological malignancies, Eur. J. Haematol. 99 kidney allograft failure: more than alloimmunity. A viewpoint article,
(4) (2017) 291–299. Transplantation 104 (2020) e46.
[5] A. Otandault, P. Anker, Z. Al Amir Dache, V. Guillaumon, R. Meddeb, B. Pastor, [30] Y. Khachatoorian, V. Khachadourian, E. Chang, E.R. Sernas, E.F. Reed, M. Deng,
et al., Recent advances in circulating nucleic acids in oncology, Ann. Oncol. 30 B.D. Piening, A.C. Pereira, B. Keating, M. Cadeiras, Noninvasive biomarkers for
(2019) 374. prediction and diagnosis of heart transplantation rejection, Transplant. Rev.
[6] S. Tug, S. Helmig, E.R. Deichmann, A. Schmeier-Jurchott, E. Wagner, T. (Orlando) 35 (1) (2021) 100590, https://doi.org/10.1016/j.trre.2020.100590.
Zimmermann, et al., Exercise-induced increases in cell free DNA in human [31] E.P. Kransdorf, M.M. Kittleson, J.K. Patel, M.J. Pando, D.E. Steidley, J.A.
plasma originate predominantly from cells of the haematopoietic lineage, Kobashigawa, Calculated panel-reactive antibody predicts outcomes on the
Exerc. Immunol. Rev. 21 (2015) 164. heart transplant waiting list, J. Heart Lung Transplant. 36 (7) (2017) 787–796.
[7] G. Barbany, C. Arthur, A. Lieden, M. Nordenskjold, R. Rosenquist, B. Tesi, et al., [32] S. Agbor-Enoh, A.M. Jackson, I. Tunc, G.J. Berry, A. Cochrane, D. Grimm, A.
Cell-free tumour DNA testing for early detection of cancer - a potential future Davis, P. Shah, A.W. Brown, Y. Wang, I. Timofte, P. Shah, S. Gorham, J. Wylie, N.
tool, J. Intern. Med. 286 (2019) 118. Goodwin, M.K. Jang, A. Marishta, K. Bhatti, U. Fideli, Y. Yang, H. Luikart, Z. Cao,
[8] A.J. Bronkhorst, V. Ungerer, F. Diehl, P. Anker, Y. Dor, M. Fleischhacker, P.B. M. Pirooznia, J. Zhu, C. Marboe, A. Iacono, S.D. Nathan, J. Orens, H.A. Valantine,
Gahan, L. Hui, S. Holdenrieder, A.R. Thierry, Towards systematic nomenclature K. Khush, Late manifestation of alloantibody-associated injury and clinical
for cell-free DNA, Hum. Genet. 140 (4) (2021) 565–578. pulmonary antibody-mediated rejection: Evidence from cell-free DNA
[9] C.K. Raymond, J. Hernandez, R. Karr, K. Hill, M. Li, F. Bertolini, Collection of cell- analysis, J. Heart Lung Transplant. 37 (7) (2018) 925–932.
free DNA for genomic analysis of solid tumors in a clinical laboratory setting, [33] H. Zhao, A. Alam, A.P. Soo, A.J.T. George, D. Ma, Ischemia-reperfusion injury
PLoS ONE 12 (4) (2017) e0176241. reduces long term renal graft survival: mechanism and beyond, EBioMedicine
[10] P. Jiang, Y.M.D. Lo, The long and short of circulating cell-free DNA and the ins 28 (2018) 31–42.
and outs of molecular diagnostics, Trends Genet. 32 (6) (2016) 360–371. [34] T.M. Snyder, K.K. Khush, H.A. Valantine, S.R. Quake, Universal noninvasive
[11] A.J. Hale, C.A. Smith, L.C. Sutherland, V.E.A. Stoneman, V.L. Longthorne, A.C. detection of solid organ transplant rejection, Proc. Natl. Acad. Sci. USA 108 (15)
Culhane, G.T. Williams, Apoptosis: molecular regulation of cell death, Eur. J. (2011) 6229–6234.
Biochem. 236 (1) (1996) 1–26. [35] International HapMap C, K.A. Frazer, D.G. Ballinger, D.R. Cox, D.A. Hinds, L.L.
[12] C. Sanchez, M.W. Snyder, R. Tanos, J. Shendure, A.R. Thierry, New insights into Stuve, et al., A second generation human haplotype map of over 3.1 million
structural features and optimal detection of circulating tumor DNA SNPs, Nature 449 (2007) 851.
determined by single-strand DNA analysis, NPJ Genom. Med. 3 (2018) 31. [36] I. De Vlaminck, H.A. Valantine, T.M. Snyder, C. Strehl, G. Cohen, H. Luikart,
[13] Y. Elshimali, H. Khaddour, M. Sarkissyan, Y. Wu, J. Vadgama, The clinical et al., Circulating cell-free DNA enables noninvasive diagnosis of heart
utilization of circulating cell free DNA (CCFDNA) in blood of cancer patients, transplant rejection, Sci. Transl. Med. 6 (2014) 241ra77.
Int. J. Mol. Sci. 14 (9) (2013) 18925–18958. [37] I. De Vlaminck, L. Martin, M. Kertesz, K. Patel, M. Kowarsky, C. Strehl, G. Cohen,
[14] Y.W. Zheng, K.C. Chan, H. Sun, P. Jiang, X. Su, E.Z. Chen, et al., H. Luikart, N.F. Neff, J. Okamoto, M.R. Nicolls, D. Cornfield, D. Weill, H.
Nonhematopoietically derived DNA is shorter than hematopoietically Valantine, K.K. Khush, S.R. Quake, Noninvasive monitoring of infection and
derived DNA in plasma: a transplantation model, Clin. Chem. 58 (2012) rejection after lung transplantation, Proc. Natl. Acad. Sci. USA 112 (43) (2015)
549. 13336–13341.
[15] H.I. Ng, X. Zhu, L. Xuan, Y. Long, Y. Mao, Y.u. Shi, L. Sun, B.o. Liang, F. Scaglia, K. [38] S. Agbor-Enoh, Y. Wang, I. Tunc, M.K. Jang, A. Davis, I. De Vlaminck, H. Luikart,
W. Choy, Z. Zhu, Analysis of fragment size distribution of cell-free DNA: A P.D. Shah, I. Timofte, A.W. Brown, A. Marishta, K. Bhatti, S. Gorham, U. Fideli, J.
potential non-invasive marker to monitor graft damage in living-related liver Wylie, D. Grimm, N. Goodwin, Y. Yang, K. Patel, J. Zhu, A. Iacono, J.B. Orens, S.D.
transplantation for inborn errors of metabolism, Mol. Genet. Metab. 127 (1) Nathan, C. Marboe, G.J. Berry, S.R. Quake, K. Khush, H.A. Valantine, Donor-
(2019) 45–50. derived cell-free DNA predicts allograft failure and mortality after lung
[16] Y.M. Lo, K.C. Chan, H. Sun, E.Z. Chen, P. Jiang, F.M. Lun, et al., Maternal plasma transplantation, EBioMedicine 40 (2019) 541–553.
DNA sequencing reveals the genome-wide genetic and mutational profile of [39] D. Zhao, T. Zhou, Y.i. Luo, C. Wu, D. Xu, C. Zhong, W. Cong, Q. Liu, J. Zhang, Q.
the fetus, Sci. Transl. Med. 2 (2010) 61ra91. Xia, Preliminary clinical experience applying donor-derived cell-free DNA to
[17] R. Meddeb, Z.A.A. Dache, S. Thezenas, A. Otandault, R. Tanos, B. Pastor, C. discern rejection in pediatric liver transplant recipients, Sci. Rep. 11 (1) (2021),
Sanchez, J. Azzi, G. Tousch, S. Azan, C. Mollevi, A. Adenis, S. El Messaoudi, P. https://doi.org/10.1038/s41598-020-80845-6.
Blache, A.R. Thierry, Quantifying circulating cell-free DNA in humans, Sci. Rep. [40] S. Agbor-Enoh, I. Tunc, I. De Vlaminck, U. Fideli, A. Davis, K. Cuttin, K.
9 (1) (2019), https://doi.org/10.1038/s41598-019-41593-4. Bhatti, A. Marishta, M.A. Solomon, A. Jackson, G. Graninger, B. Harper, H.
[18] A.S. Devonshire, A.S. Whale, A. Gutteridge, G. Jones, S. Cowen, C.A. Foy, J.F. Luikart, J. Wylie, X. Wang, G. Berry, C. Marboe, K. Khush, J. Zhu, H.
Huggett, Towards standardisation of cell-free DNA measurement in plasma: Valantine, Applying rigor and reproducibility standards to assay donor-
controls for extraction efficiency, fragment size bias and quantification, Anal. derived cell-free DNA as a non-invasive method for detection of acute
Bioanal. Chem. 406 (26) (2014) 6499–6512. rejection and graft injury after heart transplantation, J. Heart Lung
[19] K. Page, D.S. Guttery, N. Zahra, L. Primrose, S.R. Elshaw, J.H. Pringle, K. Blighe, S. Transplant. 36 (9) (2017) 1004–1012.
D. Marchese, A. Hills, L. Woodley, J. Stebbing, R.C. Coombes, J.A. Shaw, M. [41] S. Agbor-Enoh, P. Shah, I. Tunc, S. Hsu, S. Russell, E. Feller, K. Shah, M.E. Rodrigo,
Otsuka, Influence of plasma processing on recovery and analysis of circulating S.S. Najjar, H. Kong, M. Pirooznia, U. Fideli, A. Bikineyeva, A. Marishta, K. Bhatti,
nucleic acids, PLoS ONE 8 (10) (2013) e77963. Y. Yang, C. Mutebi, K. Yu, M. Kyoo Jang, C. Marboe, G.J. Berry, H.A. Valantine,
[20] F. Mauger, C. Dulary, C. Daviaud, J.-F. Deleuze, J. Tost, Comprehensive Cell-free DNA to detect heart allograft acute rejection, Circulation 143 (12)
evaluation of methods to isolate, quantify, and characterize circulating cell- (2021) 1184–1197.
free DNA from small volumes of plasma, Anal. Bioanal. Chem. 407 (22) (2015) [42] E. Sharon, H. Shi, S. Kharbanda, W. Koh, L.R. Martin, K.K. Khush, H. Valantine, J.
6873–6878. K. Pritchard, I. De Vlaminck, B.J. Raphael, Quantification of transplant-derived
[21] L. Sorber, K. Zwaenepoel, V. Deschoolmeester, G. Roeyen, F. Lardon, C. Rolfo, P. circulating cell-free DNA in absence of a donor genotype, PLoS Comput. Biol.
Pauwels, A comparison of cell-Free DNA isolation kits: isolation and 13 (8) (2017) e1005629.
quantification of cell-free DNA in plasma, J. Mol. Diagn. 19 (1) (2017) 162–168. [43] M. Grskovic, D.J. Hiller, L.A. Eubank, J.J. Sninsky, C. Christopherson, J.P. Collins,
[22] I. Alborelli, D. Generali, P. Jermann, M.R. Cappelletti, G. Ferrero, B. Scaggiante, K. Thompson, M. Song, Y.S. Wang, D. Ross, M.J. Nelles, J.P. Yee, J.C. Wilber, M.G.
M. Bortul, F. Zanconati, S. Nicolet, J. Haegele, L. Bubendorf, N. Aceto, M. Crespo-Leiro, S.L. Scott, R.N. Woodward, Validation of a clinical-grade assay to
Scaltriti, G. Mucci, L. Quagliata, G. Novelli, Cell-free DNA analysis in healthy measure donor-derived cell-free DNA in solid organ transplant recipients, J.
individuals by next-generation sequencing: a proof of concept and technical Mol. Diagn. 18 (6) (2016) 890–902.
validation study, Cell Death Dis. 10 (7) (2019), https://doi.org/10.1038/ [44] J.S. Bromberg, D.C. Brennan, E. Poggio, S. Bunnapradist, A. Langone, P. Sood, A.J.
s41419-019-1770-3. Matas, R.B. Mannon, S. Mehta, A. Sharfuddin, B. Fischbach, M. Narayanan, S.C.
[23] E.A. Bennett, D. Massilani, G. Lizzo, J. Daligault, E.M. Geigl, T. Grange, Library Jordan, D.J. Cohen, Z.S. Zaky, D. Hiller, R.N. Woodward, M. Grskovic, J.J. Sninsky,
construction for ancient genomics: single strand or double strand?, J.P. Yee, R.D. Bloom, Biological variation of donor-derived cell-free DNA in
Biotechniques 56 (2014) 289 renal transplant recipients: clinical implications, J. Appl. Lab. Med. 2 (3) (2017)
[24] A. Martuszewski, P. Paluszkiewicz, M. Król, M. Banasik, M. Kepinska, Donor- 309–321.
derived cell-free DNA in kidney transplantation as a potential rejection [45] T. Sigdel, F. Archila, T. Constantin, S. Prins, J. Liberto, I. Damm, P. Towfighi, S.
biomarker: A systematic literature review, J. Clin. Med. 10 (2) (2021) 193, Navarro, E. Kirkizlar, Z. Demko, A. Ryan, S. Sigurjonsson, R. Sarwal, S.-C. Hseish,
https://doi.org/10.3390/jcm10020193. C. Chan-On, B. Zimmermann, P. Billings, S. Moshkevich, M. Sarwal, Optimizing
[25] T. McClure, S.K. Goh, D. Cox, V. Muralidharan, A. Dobrovic, A.G. Testro, Donor- detection of kidney transplant injury by assessment of donor-derived cell-free
specific cell-free DNA as a biomarker in liver transplantation: A review, World DNA via massively multiplex PCR, J Clin Med 8 (1) (2018) 19, https://doi.org/
J. Transplant. 10 (11) (2020) 307–319. 10.3390/jcm8010019.
[26] C. Thongprayoon, P. Vaitla, I.M. Craici, N. Leeaphorn, P. Hansrivijit, S.A. Salim, T. [46] R.D. Bloom, J.S. Bromberg, E.D. Poggio, S. Bunnapradist, A.J. Langone, P. Sood, A.
Bathini, F.H. Cabeza Rivera, W. Cheungpasitporn, The use of donor-derived J. Matas, S. Mehta, R.B. Mannon, A. Sharfuddin, B. Fischbach, M. Narayanan, S.C.

857
A.M. Jackson, C. Amato-Menker and M. Bettinotti Human Immunology 82 (2021) 850–858

Jordan, D. Cohen, M.R. Weir, D. Hiller, P. Prasad, R.N. Woodward, M. Grskovic, J. [54] K.K. Khush, Clinical utility of donor-derived cell-free DNA testing in cardiac
J. Sninsky, J.P. Yee, D.C. Brennan, Cell-free DNA and active rejection in kidney transplantation, J. Heart Lung Transplant. 40 (6) (2021) 397–404.
allografts, J. Am. Soc. Nephrol. 28 (7) (2017) 2221–2232. [55] P.E. North, E. Ziegler, D.K. Mahnke, K.D. Stamm, A. Thomm, P. Daft, M. Goetsch,
[47] E.M. Gielis, K.J. Ledeganck, A. Dendooven, P. Meysman, C. Beirnaert, K. Laukens, H.L. Liang, M.A. Baker, A. Vepraskas, C. Rosenau, M. Dasgupta, P. Simpson, M.E.
et al., The use of plasma donor-derived, cell-free DNA to monitor acute Mitchell, A. Tomita-Mitchell, V. Tchantchaleishvili, Cell-free DNA donor
rejection after kidney transplantation, Nephrol. Dial. Transplant. 35 (2020) fraction analysis in pediatric and adult heart transplant patients by
714. multiplexed allele-specific quantitative PCR: Validation of a rapid and highly
[48] S.C. Jordan, S. Bunnapradist, J.S. Bromberg, A.J. Langone, D. Hiller, J.P. Yee, J.J. sensitive clinical test for stratification of rejection probability, PLoS One 15 (1)
Sninsky, R.N. Woodward, A.J. Matas, Donor-derived cell-free DNA identifies (2020) e0227385.
antibody-mediated rejection in donor specific antibody positive kidney [56] M.E. Richmond, S.D. Zangwill, S.J. Kindel, S.R. Deshpande, J.N. Schroder, D.P.
transplant recipients, Transplant. Direct 4 (9) (2018) e379, https://doi.org/ Bichell, K.R. Knecht, W.T. Mahle, M.A. Wigger, N.A. Gaglianello, E. Pahl, P.M.
10.1097/TXD.0000000000000821. Simpson, M. Dasgupta, P.E. North, M. Hidestrand, A. Tomita-Mitchell, M.E.
[49] E. Stites, D. Kumar, O. Olaitan, S. John Swanson, N. Leca, M. Weir, J. Bromberg, J. Mitchell, Donor fraction cell-free DNA and rejection in adult and pediatric
Melancon, I. Agha, H. Fattah, T. Alhamad, Y. Qazi, A. Wiseman, G. Gupta, High heart transplantation, J. Heart Lung Transplant. 39 (5) (2020) 454–463.
levels of dd-cfDNA identify patients with TCMR 1A and borderline allograft [57] D. Sayah, S.S. Weigt, A. Ramsey, A. Ardehali, J. Golden, D.J. Ross, Plasma donor-
rejection at elevated risk of graft injury, Am. J. Transplant. 20 (9) (2020) 2491– derived cell-free DNA levels are increased during acute cellular rejection after
2498. lung transplant: pilot data, Transplant. Direct 6 (2020) e608.
[50] E. Huang, S. Sethi, A. Peng, R. Najjar, J. Mirocha, M. Haas, et al., Early clinical [58] S. Tenny, M.R. Hoffman, Prevalence, StatPearls, Treasure Island (FL), 2021.
experience using donor-derived cell-free DNA to detect rejection in kidney [59] M. Oellerich, M. Shipkova, T. Asendorf, P.D. Walson, V. Schauerte, N.
transplant recipients, Am. J. Transplant. 19 (2019) 1663. Mettenmeyer, M. Kabakchiev, G. Hasche, H.-J. Gröne, T. Friede, E. Wieland, V.
[51] D.P. Puliyanda, R. Swinford, H. Pizzo, J. Garrison, A.M. De Golovine, S.C. Jordan, Schwenger, E. Schütz, J. Beck, Absolute quantification of donor-derived cell-
Donor-derived cell-free DNA (dd-cfDNA) for detection of allograft rejection in free DNA as a marker of rejection and graft injury in kidney transplantation:
pediatric kidney transplants, Pediatr. Transplant. 25 (2) (2021), https://doi.org/ Results from a prospective observational study, Am. J. Transplant. 19 (11)
10.1111/petr.v25.210.1111/petr.13850. (2019) 3087–3099.
[52] H. Zhang, C. Zheng, X. Li, Q. Fu, J. Li, Q. Su, L. Zeng, Z.u. Liu, J. Wang, H. Huang, B. [60] A.P. Cheng, P. Burnham, J.R. Lee, M.P. Cheng, M. Suthanthiran, D. Dadhania, I.
Xu, M. Ye, L. Liu, C. Wang, Diagnostic performance of donor-derived plasma De Vlaminck, A cell-free DNA metagenomic sequencing assay that integrates
cell-free DNA fraction for antibody-mediated rejection in post renal transplant the host injury response to infection, Proc. Natl. Acad. Sci. USA 116 (37) (2019)
recipients: A prospective observational study, Front. Immunol. 11 (2020), 18738–18744.
https://doi.org/10.3389/fimmu.2020.00342. [61] F. Erger, D. Norling, D. Borchert, E. Leenen, S. Habbig, M.S. Wiesener, et al.,
[53] J. Shen, L. Guo, P. Yan, J. Zhou, Q. Zhou, W. Lei, H. Liu, G. Liu, J. Lv, F. Liu, H. cfNOMe - A single assay for comprehensive epigenetic analyses of cell-free
Huang, W. Dong, L. Shu, H. Wang, J. Wu, J. Chen, R. Wang, Prognostic value of DNA, Genome Med. 12 (2020) 54.
the donor-derived cell-free DNA assay in acute renal rejection therapy: A [62] M.W. Snyder, M. Kircher, A.J. Hill, R.M. Daza, J. Shendure, Cell-free DNA
prospective cohort study, Clin. Transplant. 34 (10) (2020), https://doi.org/ comprises an in vivo nucleosome footprint that informs its tissues-of-origin,
10.1111/ctr.v34.1010.1111/ctr.14053. Cell 164 (2016) 57.

858

You might also like