Next Article in Journal
Persufflation—Current State of Play
Next Article in Special Issue
Animal Models in Allogenic Solid Organ Transplantation
Previous Article in Journal
Epitope-Level Matching—A Review of the Novel Concept of Eplets in Transplant Histocompatibility
Previous Article in Special Issue
The Effects of 6-Chromanol SUL-138 during Hypothermic Machine Perfusion on Porcine Deceased Donor Kidneys
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Donor-Derived Cell-Free DNA to Diagnose Graft Rejection Post-Transplant: Past, Present and Future

1
Department of Anthropology, University of Delhi, Delhi 110007, India
2
Department of Research, Sir Ganga Ram Hospital, Delhi 110060, India
3
Department of Nephrology, Sir Ganga Ram Hospital, Delhi 110060, India
*
Author to whom correspondence should be addressed.
Transplantology 2021, 2(3), 348-361; https://doi.org/10.3390/transplantology2030034
Submission received: 17 July 2021 / Revised: 14 August 2021 / Accepted: 8 September 2021 / Published: 10 September 2021

Abstract

:
Donor-derived cell-free DNA (dd-cfDNA) is a non-invasive biomarker that is more sensitive and specific towards diagnosing any graft injury or rejection. Due to its applicability over all transplanted organs irrespective of age, sex, race, ethnicity, and the non-requirement of a donor sample, it emerges as a new gold standard for graft health and rejection monitoring. Published research articles describing the role and efficiency of dd-cfDNA were identified and scrutinized to acquire a brief understanding of the history, evolution, emergence, role, efficiency, and applicability of dd-cfDNA in the field of transplantation. The dd-cfDNA can be quantified using quantitative PCR, next-generation sequencing, and droplet digital PCR, and there is a commendatory outcome in terms of diagnosing graft injury and monitoring graft health. The increased levels of dd-cfDNA can diagnose the rejection prior to any other presently used biochemistry or immunological assay methods. Biopsies are performed when these tests show any signs of injury and/or rejection. Therefore, by the time these tests predict and show any unusual or improper activity of the graft, the graft is already damaged by almost 50%. This review elucidates the evolution, physiology, techniques, limitations, and prospects of dd-cfDNA as a biomarker for post-transplant graft damage and rejection.

1. Introduction

Solid organ transplant (SOT) is an effective therapy for end-stage diseases and organ failures, which is a lifesaving, transformational and restorative procedure for improving the quality and longevity of life. SOT relies on the effective surveillance of graft which includes serial laboratory testing and biopsies.
163,138 transplants were performed in 2019 worldwide, including 105,231 kidney transplants, 39,007 liver transplants, 9140 heart transplants, 7144 lung transplants, 2479 pancreas transplants, and 137 small bowel transplants [1]. Every transplant needs precise and frequent monitoring. Biopsies cannot be performed frequently and are performed when any clinical symptoms are triggered [2,3].
The immune system acts on all foreign particles that enter the body and produces antibodies in response. Similarly, when an organ is transplanted, the immune system recognizes it and acts against it, consequently destroying the perceived foreign cells in the form of the transplanted organ [4,5]. Due to the huge gap between the need and availability of organs, one cannot risk an organ failure after a transplant, owing merely to the immune response. To overcome this several measures are taken at each step, right from the beginning where the best suitable match for the organ is searched for, succeeded by the advanced medications and immunotherapy given to the recipients in order to suppress the immune response, until the enhanced post-transplant patient care.
Post-transplant recipients are kept under surveillance and strict regimes to avoid any complications that may lead to either damage or rejection of the transplanted organ [6,7]. Around 7% of renal transplants fail after a year, 17% fail after 3 years, and 46% fail after 10 years [8]. Among heart transplants, about 10% of the transplants fail in one year and 30% in 5 years [9,10,11]. Similarly, among liver transplant recipients around 20% of failures are reported in 1 year and 30% of failures in 5 years [12,13], thereby making graft rejection a pertinent obstacle in transplantation.
Large-scale screening processes undertaken in the 2000s in Australia, Norway, and the United States revealed that more than 10% of adults had markers for chronic kidney disease (CKD). The prevalence of CKD is in 9.1% of the global population [14]. More than 500,000 people in the United States live with ESRD [15]. Heart failure is a global health issue, with a prevalence of over 23 million cases worldwide [16]. It is predicted that by the end of 2030; 3 million additional patients will be added to these figures [17]. One out of every nine deaths in America is caused by heart failure [18]. There were 112 million reported cases of prevalent compensated cirrhosis and 10.2 million cases of decompensated cirrhosis worldwide, in 2017 [19]. The figures are indicative of the rising cases of organ failures and thereby hint at a scenario of unaffordable graft rejections.
After transplantation patients are required to undergo biopsy in order to diagnose graft dysfunction or rejection. Therefore, there is a pressing need to explore potential biomarkers which could be used as early detection mechanisms for monitoring graft health, graft damage, and rejection. The currently available non-invasive methods to detect damage and rejection are insufficient and donor-derived cell-free DNA (dd-cfDNA) seems to be very promising as a non-invasive biomarker. This review was conducted to explore the evolution of dd-cfDNA, its utility and future perspectives as a potential biomarker, as it summarizes the origin, current advancements and efficacy of dd-cfDNA in diagnosing graft injury and rejection. We believe that this review will inspire researchers to carry out further research in the field and add to the existing body of knowledge.

2. Materials and Methods

2.1. Search Strategy

This review was conducted in accordance with Preferred Reporting Items for Systematic and Meta-Analysis (PRISMA) guidelines. A literature review was performed using PubMed and Web of Science for finding relevant studies published as of 31 January 2021, without any language restrictions. The full search strategy is depicted in Figure 1.
The search terms included “donor derived cell free DNA”, “dd-cfDNA” and “graft cell free DNA”. Relevant literature reviews and references of included studies were also searched to identify other relevant analyses.

2.2. Study Selection

We included original articles published up to 31 January 2021 that investigated the role and utility of donor-derived cell-free DNA in diagnosing or monitoring graft rejection or graft health. We considered all types of study-randomized control trials, non-randomized prospective cohorts, retrospective cohorts, case reports, or case series. Inclusion and exclusion criteria were formulated to ensure comprehensive searching and screening for published articles relevant to diagnosing rejection/health of any solid organ transplant. Studies were excluded if they met any of the following criteria: markers other than donor-derived cell-free DNA, transplants other than solid organs, non-English publication, unavailable full texts, or non-peer reviewed texts. The published study reporting the utility of dd-cfDNA in graft health monitoring and rejection diagnosis were included.
Titles and abstracts were independently reviewed by two authors (N.K. and R.R.). After screening, the full text of the articles was reviewed by three authors (N.K., R.R. and A.G.). The two screeners discussed items in which there was a disagreement and if a consensus could not be reached, the third author’s adjudication was sought (M.P.S.).

3. Evolution of Analytical Approaches and Techniques to Diagnose Graft Rejection Using dd-cfDNA

Thomas Earl Starzl, an American physician who performed the first liver transplant in humans, provided the first ever well-defined evidence for the presence of donor DNA in the bloodstream of a recipient, which was ascertained by karyotyping studies in female recipients of liver transplants who obtained a graft from cadaveric male donors and proved that the organ genome becomes chimera in transplanted patients [20]. ‘Graft chimerism might be a generic feature of all accepted graft’ was theorized after the evidence of chimeras observed in intestine, kidney, lung, heart, and liver transplants [21,22,23,24,25].
Evidence of the presence of Y chromosomes in the recipient’s urine and plasma of sex-mismatched donor-recipient pairs revealed that increased level of dd-cfDNA could be an indicator for rejection and acceptance of the graft [26,27,28,29,30]. A major drawback of this approach was that it was pertinent only to a female recipient who obtained an organ from a male donor. Therefore, this limited the number of transplant cases and reduced its applicability in a minority of cases. Despite its limitations, it led to the genesis of the idea of microchimerism, which is the presence of small amounts of cells originated from another individual having different genetic information from the host.
This microchimerism directed the researchers and scientists to identify and quantify the donor DNA in the recipient by discovering an assay which could be applicable to all the recipients irrespective of age, sex and the type of organ transplanted. Hence a universal, sex-independent assay was introduced which required the genotyping of both donor and recipient. Single nucleotide polymorphisms (SNP), which are homozygous and can distinguish donor DNA from recipient DNA, were selected for further analysis and quantification. This method relied upon knowledge of donor and recipient genotypes, which required time, a donor sample, and was costly. Shotgun sequencing was not possible in a regular hospital setting because of its high turnaround time and because genotyping both the donor and recipient was time- and effort-constraining.
Thus, the proposed technique takes a leap over the need of genotyping both the donor and recipient as it proposes the uses of the assay of preselected SNPs which are applicable to all donor recipients’ pairs, irrespective of age, sex, ethnicity, and organs. An SNP should be homozygous in both donor and recipient, and should differ between the two. To select such SNPs, we can either genotype both donors and recipients and select the homozygous different SNPs which identify donor DNA and can be further quantified. The other approach is preselecting a panel of informative SNPs from the public genomic databases like HapMap, 1000 Genomes, and Indi Genomes. This informative panel of preselected SNPs contains SNPs with high minor allele frequency (MAF) of 0.4–0.5. The SNPs selected should not be located within or directly adjacent to an annotated repetitive element. Considering the Hardy–Weinberg equilibrium an SNP with an MAF of 0.4–0.5 has a nearly equal distribution of both alleles in a population and an 11.5 to 12.5% chance of containing a homozygous genotype in two individuals: the recipient and the donor. It has approximately a 23–25% chance of being homozygous for each allele in an individual. Hence, an informative panel of a set of 30–35 SNPs is enough to identify 3 SNPs for each donor–recipient pair to assess the damage and rejection in the recipient [31]. However, if SNPs were selected randomly without the required MAF, then approximately 3000 SNPs would be available to test for homozygosity between the donor and the recipient [32].
Presently, there are two simultaneous techniques for using dd-cfDNA; as biomarkers, based on the measurement of the number of donor molecules by next-generation sequencing or the digital PCR method. The next-generation sequencing method uses 266 SNPs panel in comparison to 41 SNPs used by the droplet digital method by Beck et al. [32,33]. Both of these methods lead to the same results [34].
An individual genome carries approximately 4–5 million SNPs and there are more than 100 million SNPs across the populations globally. Only 0.1–1% of DNA differ between any two individuals, depending on the degree of relatedness [35]. An SNP may be unique to an individual or a population. In forensic science, SNPs are used to establish identities. Similarly, the SNPs are used in transplantation for identifying different DNA circulating in the plasma of the recipient.
Genomic biomarkers are more complex than standard chemistry diagnostics. Hence, they require advanced methods, workflows, and analytical approaches. The assays detecting dd-cfDNA differ in technology and methods. Therefore, each of the assays requires clinical validation before introducing it in a clinical setting. The chronology of the advancement and use of different methods and techniques for identifying and quantifying the donor-derived cell-free DNA during the last two decades are elucidated in Table 1.

4. The dd-cfDNA as a Potential Biomarker

Donor-derived cell-free DNA (dd-cfDNA), also referred to as graft cell-free DNA (GcfDNA) or donor-specific DNA (ds-DNA), is a biomarker which works on the principle/fact that an organ transplant is not just a transplant of an organ but also the transplant of a genome. The biological rationale behind the utility of dd-cfDNA is that the immune system is activated upon recognition of the transplanted organ and produces antibodies in response. The antibodies attack the transplanted organ which leads to apoptosis or cell necrosis. The ruptured or dead cells release their contents into the recipient’s blood plasma and thereafter, the recipient carries the DNA of the donor, which reflects in its name, ‘donor-derived cell-free DNA’. This is cell-free, circulating and donor DNA.
Donor-derived cell-free DNA is found in all recipients irrespective of whether the recipient shows rejection or not, although the number of cells may vary from individual to individual (Figure 2) [20]. Furthermore, even after using different methods for the identification and quantification of dd-cfDNA, in stable patients similar median values are obtained [61].
Large numbers of donor cells are present in individuals experiencing graft damage and/or rejection due to the breakdown of cells.
The clearance of dd-cfDNA from an individual’s body is similar to that of cell-free DNA, though it is still being studied and requires further research.
The half-life of cell-free DNA in the bloodstream is between 16 min to 2.5 h [62,63]. Cell-free DNA can enter the liver or spleen and be degraded by the macrophages that are located inside by the action of the DNase I enzyme [64]. Cell-free DNA can also be excreted through urine.
The pertinent questions that open the purview of the discussion concerning “whether dd-cfDNA is cleared out from the body through urine?” and “Is increased level of dd-cfDNA associated with impaired kidney function rather than actual graft damage?” Some insight can be gained even though it is unknown which mechanism is involved in clearing out the cell-free DNA from the blood plasma of the recipient, but a study on foetal DNA cleared out from maternal blood circulation showed that only 0.2 to 19% of the total foetal DNA is cleared out by the kidney, stating that renal excretion is not a major route when it comes to the clearance of non-host (foetal) DNA [65]. Donor DNA is always present in the plasma of the recipient in varying quantities.
The recipient also carries its own cell-free DNA. So, the quantification of donor DNA will help to monitor the graft health, damage and rejection. For the quantification of dd-cfDNA, SNPs are best-suited as they can differentiate dd-cfDNA from the recipient cf-DNA. The polymorphism between the donor and the recipient must be identified. Therefore, for diagnosing graft damage and rejection, the panel of SNPs used must be able to differentiate between individuals.
Currently used methods for monitoring the organ health and diagnosing the rejection are based on signs and symptoms of patient, biochemistry tests, immunological assays, and biopsies. Serum creatinine (sCr) is increased if there is a significant reduction in the glomerular filtration rate (GFR), and about 50% of the kidney function is lost before a rise in sCr is detected, hence making it a late marker for acute rejection [66].
Biopsy being a ‘gold standard’ for assessing the health and rejection of an organ is an invasive technique and has its own set of limitations, including the turnaround time, expense, inconvenient process, chances of sampling error, inter-observer variability and poor recommendation for serial testing. Biopsies are performed on suspicion of damage or graft rejection and almost 50% or more of the organ is typically injured before it is recommended for biopsy [66,67,68]. Due to this, asymptomatic graft injuries remain undiagnosed. However, endomyocardial biopsy is also not a true gold standard for diagnosing acute rejection [50].
Approximately 25% of biopsies yield an inadequate specimen and this risk can become larger with a smaller needle size. Because of the modern immunosuppression, many units no longer perform routine biopsies [69]. In a study by Bloom et al. it was found that only 27% of the clinically indicated biopsies in kidney transplant patients revealed active rejection, where the biopsies were performed after observing an elevation in the sCr over baseline [36]. In another study by Yang et al., 43% of the clinically indicated biopsies were found quiescent and 65% of the protocol biopsies were tagged as unnecessary invasive procedures [4].

5. Monitoring of Organ Damage and Rejection through dd-cfDNA

The monitoring of dd-cfDNA is based on the number of donor DNA copies circulating in the plasma of recipients. The levels of dd-cfDNA in stable patients are significantly less when compared to rejection cases. High levels of dd-cfDNA are associated with the rejection cases in liver, kidney, heart, lung transplant recipients [27,28,29,30,32,33,34,36,37,38,39,40,42,43,44,47,48,49,50,52,53,70,71,72]. The dd-cfDNA levels begin to rise from weeks to months before acute rejection is diagnosed by the currently used methods. These increased levels represent early graft damage or injury [3,30,36]. The dd-cfDNA is highly efficient in detecting injury and rejection caused due to antibody mediated rejection (ABMR). But, when it comes to T-cell mediated rejection (TCMR), some studies have also shown that TCMR is associated with elevation in dd-cfDNA. TCMR type IB, type IIA, type IA and borderline rejection shows a high percentage of dd-cfDNA, but according to a study by Sigdel et al., it is averred that the dd-cfDNA method is unable to detect TCMR [45,52,53,59]. Further studies are needed to assess the role of dd-cfDNA in detecting TCMR rejection.
The elevation of dd-cfDNA is not always linked to rejection but also points out the degree of graft injury and infection. Immediately post-transplant the level of dd-cfDNA is increased but decreases after a couple of days of kidney transplant, liver transplant, lung transplant and heart transplant, which is also reflective of ischemia or reperfusion injuries in the transplanted organs (Figure 3) [26,32,39,41,47,48].
The levels of dd-cfDNA are affected by anti-rejection therapy and are dose-dependent. Levels of dd-cfDNA decrease rapidly after anti-rejection therapy [57]. This paves the way for us to create personalized immunosuppressive drugs to prevent graft damage and rejection. Furthermore, this will also help to minimize the risk of other diseases, caused by the suppressed immune system of the body and high dosage of immunosuppressive medicines.
It is reported through 16 studies upon the utility and effectiveness of dd-cfDNA in detecting rejection among kidney, heart, liver and lung transplants that the mean sensitivities and specificities of dd-cfDNA are 79.83 ± 9.15% and 79.61 ± 14.86%, respectively (see Table 2). These studies detail the efficiency of dd-cfDNA to detect antibody-mediated rejection (ABMR) primarily.

6. Limitations of dd-cfDNA as a Potential Biomarker

The dd-cfDNA method cannot be applied to transplants between identical twins because of the similar genetic makeup which will make it impossible to differentiate donor and recipient DNA [33]. In the case of dual organ transplant from a single donor, dd-cfDNA can identify graft injury but cannot distinguish the affected organ. Furthermore, this will not be applicable to multi-organ transplant recipients where there is more than one donor involved, as the donor DNA will be variable and the recipient will have cell-free donor DNA from more than one graft. Thus, it will be difficult to assess which graft is subject to injury and at a risk of rejection. If one must monitor and study donor DNA in a patient with multiple organ transplants, then one would have to perform genotyping on the recipient’s and donor’s DNA to assess the status of rejection of the organ [33].
Since dd-cfDNA increases with graft injury, the impact of the BK virus, nephrotic syndrome or urinary tract infection on dd-cfDNA levels in kidney transplant patients is still unknown and requires further research.
Lastly, studies directly comparing different techniques and approaches for identifying and quantifying dd-cfDNA are unavailable. Each new assay should require clinical validation before applicability.

7. Conclusions

An understanding of the methods and techniques used to diagnose rejection and graft damage through dd-cfDNA will allow clinicians to understand its applicability and utility in clinical settings. Several studies have clinically validated the utility of dd-cfDNA in monitoring graft damage and diagnosing rejection.
The dd-cfDNA has the advantage of not being affected by any pathological or bodily functions and interrogates the health of graft directly. It allows for the early detection of rejection episodes which will enable clinicians to quickly intervene with immunosuppressive therapies and prevent graft damage or failure. Since it is a non-invasive method, it can be performed frequently and thus paves the way for personalized immunosuppressive therapies. This will prevent recipients from under- or over-immunosuppression and help in minimizing the side effects of immunosuppressive therapies.
The dd-cfDNA may compliment or replace the post-transplant methods to diagnose graft health and rejection. It can help in avoiding unnecessary biopsies. A graft rejection monitoring technique in the form of dd-cfDNA, also referred to as liquid biopsy, opens a new vista for becoming an upcoming gold standard.

8. Future Prospect of dd-cfDNA

The dd-cfDNA has a highly favorable diagnostic performance for graft rejection or injury. It has shown the evidence towards the detection of graft injury and rejection before it can be diagnosed by the present diagnostic methods. It will guide surgeons and transplant teams so that they have enough time to act and prevent graft injury and, in the worst-case scenario, graft loss. The high negative predictive value can be used to avoid unnecessary biopsies. It will help in reducing the risk and discomfort of biopsy along with reducing the burden of cost on the recipient, dd-cfDNA detects the graft injury by immune attack, which is caused by the activation of the immune system, indicating the insufficiency of immunosuppression medications, therefore indicating its potential to help in monitoring immunosuppression and bringing to light the aspect of individualized dosage. This will further help medical professionals to lower the chance of rejection, infection, and other side effects of immunosuppression. This diagnostic method can provide patients with an option to get diagnosed from a home sample collection. This not only makes the diagnosis convenient for patients but can also be used during the COVID-19 pandemic. Transplant patients are at a high risk of contracting COVID-19, and even dying from the disease due to a compromised immune system [76]. Therefore, home sample collection allows the recipient a safer option for receiving a diagnosis along with tele-consultation. Therefore, serial dd-cfDNA monitoring will lead to the cost-effective personalized management of transplant recipients to reduce graft loss.
Future studies should include large cohorts with different dd-cfDNA diagnostic approaches and techniques along with clinical outcomes, as well as the personalization of immunosuppression. The impact of other diseases and pathophysiology also needs to be investigated.

Author Contributions

N.K., R.R., and A.G. contributed to the acquisition of data, analysis and interpretation of data, and drafted the article; D.S.R. and M.P.S. aided in revising the article and including critically important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

N.K. is receiving doctoral fellowship from Indian Council of Social Science Research [RFD/2019-20/GEN/HLTH/177].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All articles are available from PubMed and Web of Science.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Global Observatory on Donation and Transplantation. Available online: http://www.transplant-observatory.org/data-charts-and-tables/ (accessed on 27 February 2021).
  2. Beck, J.; Oellerich, M.; Schutz, E. A New Universal Multiplex Digital PCR Method with Improve Precision for the Quantification of Donor Derived graft cfDNA Traces. Available online: http://chronixbiomedical.com/wp-content/uploads/2018/08/transplant-Beck-AACC-2015-2-copy.pdf (accessed on 20 March 2020).
  3. Schütz, E.; Fischer, A.; Beck, J.; Harden, M.; Koch, M.; Wuensch, T.; Stockmann, M.; Nashan, B.; Kollmar, O.; Matthaei, J.; et al. Graft-derived cell-free DNA, a noninvasive early rejection and graft damage marker in liver transplantation: A prospective, observational, multicenter cohort study. PLoS Med. 2017, 14, e1002286. [Google Scholar] [CrossRef] [PubMed]
  4. Yang, J.Y.; Sarwal, R.D.; Sigdel, T.K.; Damm, I.; Rosenbaum, B.; Liberto, J.M.; Chan-On, C.; Arreola-Guerra, J.M.; Alberu, J.; Vincenti, F.; et al. A urine score for non-invasive accurate diagnosis and prediction of kidney transplant rejection. Sci. Transl. Med. 2020, 12, eaba2501. [Google Scholar] [CrossRef]
  5. Alraies, M.C.; Eckman, P. Adult heart transplant: Indications and outcomes. J. Thorac. Dis. 2014, 6, 1120. [Google Scholar] [PubMed]
  6. National Kidney Foundation. Steps to Keep Your Transplanted Kidney. Available online: https://www.kidney.org/atoz/content/keepyourtransplantedkidney (accessed on 2 January 2021).
  7. Mayo Clinic. Liver Transplant. Available online: https://www.mayoclinic.org/tests-procedures/liver-transplant/about/pac-20384842 (accessed on 24 December 2020).
  8. National Kidney Foundation. Dialysis. Available online: https://www.kidney.org/atoz/content/dialysisinfo/ (accessed on 30 October 2020).
  9. Russo, M.J.; Rana, A.; Chen, J.M.; Hong, K.N.; Gelijns, A.; Moskowitz, A.; Widmann, W.D.; Ratner, L.; Naka, Y.; Hardy, M.A. Pretransplantation patient characteristics and survival following combined heart and kidney transplantation: An analysis of the United Network for Organ Sharing Database. Arch. Surg. 2009, 144, 241–246. [Google Scholar] [CrossRef] [Green Version]
  10. Russo, M.J.; Chen, J.M.; Hong, K.N.; Stewart, A.S.; Ascheim, D.D.; Argenziano, M.; Mancini, D.M.; Oz, M.C.; Naka, Y. Columbia University Heart Transplant Outcomes Research Group. Survival after heart transplantation is not diminished among recipients with uncomplicated diabetes mellitus. Circulation 2006, 114, 2280–2287. [Google Scholar] [CrossRef] [Green Version]
  11. Russo, M.J.; Davies, R.R.; Sorabella, R.A.; Martens, T.P.; George, I.; Cheema, F.H.; Mital, S.; Mosca, R.S.; Chen, J.M. Adult-age donors offer acceptable long-term survival to pediatric heart transplant recipients: An analysis of the United Network of Organ Sharing database. J. Thorac. Cardiovasc. Surg. 2006, 132, 1208–1212. [Google Scholar] [CrossRef] [Green Version]
  12. Chu, K.K.; Chan, S.C.; Sharr, W.W.; Chok, K.S.; Dai, W.C.; Lo, C.M. Low-volume deceased donor liver transplantation alongside a strong living donor liver transplantation service. World J. Surg. 2014, 38, 1522–1528. [Google Scholar] [CrossRef] [PubMed]
  13. Jones, P.D.; Hayashi, P.H.; Barritt, A. Liver transplantation in 2013: Challenges and controversies. Minerva Gastroenterol. 2013, 59, 117–131. [Google Scholar]
  14. Bikbov, B.; Purcell, C.A.; Levey, A.S.; Smith, M.; Abdoli, A.; Abebe, M.; Adebayo, O.M.; Afarideh, M.; Agarwal, S.K.; Agudelo-Botero, M.; et al. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef] [Green Version]
  15. Benjamin, O.; Lappin, S.L. End-stage renal disease. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA. Available online: https://www.ncbi.nlm.nih.gov/books/NBK499861/ (accessed on 25 March 2020).
  16. Bui, A.L.; Horwich, T.B.; Fonarow, G.C. Epidemiology and risk profile of heart failure. Nat. Rev. Cardiol. 2011, 8, 30. [Google Scholar] [CrossRef] [Green Version]
  17. Roger, V.L.; Weston, S.A.; Redfield, M.M.; Hellermann-Homan, J.P.; Killian, J.; Yawn, B.P.; Jacobsen, S.J. Trends in heart failure incidence and survival in a community-based population. JAMA 2004, 292, 344–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Go, A.S.; Mozaffarian, D.; Roger, V.L.; Benjamin, E.J.; Berry, J.D.; Borden, W.B.; Bravata, D.M.; Dai, S.; Ford, E.S.; Fox, C.S.; et al. Heart disease and stroke statistics—2013 update: A report from the American Heart Association. Circulation 2013, 127, e6–e245. [Google Scholar] [CrossRef]
  19. Sepanlou, S.G.; Safiri, S.; Bisignano, C.; Ikuta, K.S.; Merat, S.; Saberifiroozi, M.; Poustchi, H.; Tsoi, D.; Colombara, D.V.; Abdoli, A.; et al. The global, regional, and national burden of cirrhosis by cause in 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 2020, 5, 245–266. [Google Scholar] [CrossRef] [Green Version]
  20. Starzl, T.E.; Putnam, C.W. Experience in Hepatic Transplantation; WB Saunders Company: Philadelphia, PA, USA, 1969. [Google Scholar]
  21. Iwalrl, Y.; Starzl, T.E.; Yagihashi, A. Replacement of donor lymphoid tissue in human small bowel transplants under FK 506 immunosuppression. Lancet 1991, 337, 818. [Google Scholar]
  22. Startzl, T.E.; Dimetris, A.J.; Trucco, M. Chimerism and donorspecific nonreactivity 27–29 years after kidney transplantation. Transplantation 1993, 55, 1272–1277. [Google Scholar] [CrossRef] [Green Version]
  23. Fung, J.J.; Zeevi, A.; Kaufman, C.; Paradis, I.L.; Dauber, J.H.; Hardesty, R.L.; Griffith, B.; Duquesnoy, R.J. Interactions between bronchoalveolar lymphocytes and macrophages in heart-lung transplant recipients. Hum. Immunol. 1985, 14, 287–294. [Google Scholar] [CrossRef]
  24. Demtris, A.J.; Murase, N.; Starzl, T.E. Donor dendritic cells in graft and host lymphoid and non-lymphoid tissues after liver and heart allotransplantation under short term immunosuppression. Lancet 1992, 339, 1610. [Google Scholar] [CrossRef] [Green Version]
  25. Valdivia, L.A.; Demetris, A.J.; Langer, A.M.; Celli, S.; Fung, J.J.; Starzl, T.E. Dendritic cell replacement in long-surviving liver and cardiac xenografts. Transplantation 1993, 56, 482. [Google Scholar] [CrossRef] [Green Version]
  26. Zhang, J.; Tong, K.L.; Li, P.K.; Chan, A.Y.; Yeung, C.K.; Pang, C.C.; Wong, T.Y.; Lee, K.C.; Lo, Y.D. Presence of donor-and recipient-derived DNA in cell-free urine samples of renal transplantation recipients: Urinary DNA chimerism. Clin. Chem. 1999, 45, 1741–1746. [Google Scholar] [CrossRef]
  27. García Moreira, V.; Prieto García, B.; Baltar Martín, J.M.; Ortega Suárez, F.; Alvarez, F.V. Cell-free DNA as a noninvasive acute rejection marker in renal transplantation. Clin. Chem. 2009, 55, 1958–1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Macher, H.C.; Suárez-Artacho, G.; Guerrero, J.M.; Gómez-Bravo, M.A.; Álvarez-Gómez, S.; Bernal-Bellido, C.; Dominguez-Pascual, I.; Rubio, A. Monitoring of transplanted liver health by quantification of organ-specific genomic marker in circulating DNA from receptor. PLoS ONE 2014, 9, e113987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Sigdel, T.K.; Vitalone, M.J.; Tran, T.Q.; Dai, H.; Hsieh, S.C.; Salvatierra, O.; Sarwal, M.M. A rapid noninvasive assay for the detection of renal transplant injury. Transplantation 2013, 96, 97. [Google Scholar] [CrossRef] [Green Version]
  30. Snyder, T.M.; Khush, K.K.; Valantine, H.A.; Quake, S.R. Universal noninvasive detection of solid organ transplant rejection. Proc. Natl. Acad. Sci. USA 2011, 108, 6229–6234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Beck, J.; Oellerich, M.; Schütz, E. A Universal Droplet Digital PCR Approach for Monitoring of Graft Health after Transplantation Using a Preselected SNP Set; Digital PCR; Humana Press: New York, NY, USA, 2018; pp. 335–348. [Google Scholar]
  32. Beck, J.; Bierau, S.; Balzer, S.; Andag, R.; Kanzow, P.; Schmitz, J.; Gaedcke, J.; Moerer, O.; Slotta, J.E.; Walson, P.; et al. Digital droplet PCR for rapid quantification of donor DNA in the circulation of transplant recipients as a potential universal biomarker of graft injury. Clin. Chem. 2013, 59, 1732–1741. [Google Scholar] [CrossRef] [Green Version]
  33. Grskovic, M.; Hiller, D.J.; Eubank, L.A.; Sninsky, J.J.; Christopherson, C.; Collins, J.P.; Thompson, K.; Song, M.; Wang, Y.S.; Ross, D.; et al. Validation of a clinical-grade assay to measure donor-derived cell-free DNA in solid organ transplant recipients. J. Mol. Diagn. 2016, 18, 890–902. [Google Scholar] [CrossRef] [Green Version]
  34. Weir, M.; Hiller, D.; Yee, J.; Matas, A. Donor-derived cell-free DNA outperforms serum creatinine changes for identifying kidney transplant rejection. Am. J. Transplant. 2018, 18 (Suppl. 4), 506. [Google Scholar]
  35. Consortium, G.P. A global reference for human genetic variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef] [Green Version]
  36. Grskovic, M.; Hiller, D.J.; Eubank, L.A.; Sninsky, J.J.; Christopherson, C.; Collins, J.P.; Thompson, K.; Song, M.; Wang, Y.S.; Ross, D.; et al. Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection. Sci. Transl. Med. 2014, 6, 241ra77. [Google Scholar]
  37. De Vlaminck, I.; Martin, L.; Kertesz, M.; Patel, K.; Kowarsky, M.; Strehl, C.; Cohen, G.; Luikart, H.; Neff, N.F.; Okamoto, J.; et al. Noninvasive monitoring of infection and rejection after lung transplantation. Proc. Natl. Acad. Sci. USA 2015, 112, 13336–13341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Beck, J.; Oellerich, M.; Schulz, U.; Schauerte, V.; Reinhard, L.; Fuchs, U.; Knabbe, C.; Zittermann, A.; Olbricht, C.; Gummert, J.F.; et al. Donor-derived cell-free DNA is a novel universal biomarker for allograft rejection in solid organ transplantation. Transplant. Proc. 2015, 47, 2400–2403. [Google Scholar] [CrossRef] [PubMed]
  39. Burnham, P.; Kim, M.S.; Agbor-Enoh, S.; Luikart, H.; Valantine, H.A.; Khush, K.K.; De Vlaminck, I. Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cell-free DNA in plasma. Sci. Rep. 2016, 6, 1–9. [Google Scholar]
  40. Macher, H.C.; Suárez-Artacho, G.; Jiménez-Arriscado, P.; Álvarez-Gómez, S.; García-Fernández, N.; Guerrero, J.M.; Molinero, P.; Trujillo-Arribas, E.; Gómez-Bravo, M.A.; Rubio, A. Evaluation of the state of transplanted liver health by monitoring of organ-specific genomic marker in circulating DNA from receptor. Adv. Exp. Med. Biol. 2016, 924, 113–116. [Google Scholar] [PubMed]
  41. Schutz, E.; Blum, A.; Beck, J.; Harden, M.; Koch, M.; Wuensch, T.; Stockmann, M.; Nashan, B.; Kollmar, O.; Matthaei, J.; et al. Graft-derived cell-free DNA-a promising rejection marker in liver transplantation-results from a prospective multicenter trial. Clin. Chem. 2016, 62, S41. [Google Scholar]
  42. Zou, J.; Duffy, B.; Slade, M.; Young, A.L.; Steward, N.; Hachem, R.; Mohanakumar, T. Rapid detection of donor cell-free DNA in lung transplant recipients with rejections using donor-recipient HLA mismatch. Hum. Immunol. 2017, 78, 342–349. [Google Scholar] [CrossRef] [Green Version]
  43. Bromberg, J.S.; Brennan, D.C.; Poggio, E.; Bunnapradist, S.; Langone, A.; Sood, P.; Matas, A.J.; Mannon, R.B.; Mehta, S.; Sharfuddin, A.; et al. Biological variation of donor-derived cell-free DNA in renal transplant recipients: Clinical implications. J. Appl. Lab. Med. 2017, 2, 309–321. [Google Scholar] [CrossRef] [Green Version]
  44. Goh, S.K.; Muralidharan, V.; Christophi, C.; Do, H.; Dobrovic, A. Probe-free digital PCR quantitative methodology to measure donor-specific cell-free DNA after solid-organ transplantation. Clin. Chem. 2017, 63, 742–750. [Google Scholar] [CrossRef] [Green Version]
  45. Bloom, R.D.; Bromberg, J.S.; Poggio, E.D.; Bunnapradist, S.; Langone, A.J.; Sood, P.; Matas, A.J.; Mehta, S.; Mannon, R.B.; Sharfuddin, A.; et al. Cell-free DNA and active rejection in kidney allografts. J. Am. Soc. Nephrol. 2017, 28, 2221–2232. [Google Scholar] [CrossRef]
  46. Tanaka, S.; Sugimoto, S.; Kurosaki, T.; Miyoshi, K.; Otani, S.; Suzawa, K.; Hashida, S.; Yamane, M.; Oto, T.; Toyooka, S. Donor-derived cell-free DNA is associated with acute rejection and decreased oxygenation in primary graft dysfunction after living donor-lobar lung transplantation. Sci. Rep. 2018, 8, 1–9. [Google Scholar] [CrossRef]
  47. Jordan, S.C.; Bunnapradist, S.; Bromberg, J.S.; Langone, A.J.; Hiller, D.; Yee, J.P.; Sninsky, J.J.; Woodward, R.N.; Matas, A.J. Donor-derived cell-free DNA identifies antibody-mediated rejection in donor specific antibody positive kidney transplant recipients. Transplant. Direct. 2018, 4, e379. [Google Scholar] [CrossRef]
  48. Gielis, E.M.; Beirnaert, C.; Dendooven, A.; Meysman, P.; Laukens, K.; De Schrijver, J.; Van Laecke, S.; Van Biesen, W.; Emonds, M.P.; De Winter, B.Y.; et al. Plasma donor-derived cell-free DNA kinetics after kidney transplantation using a single tube multiplex PCR assay. PLoS ONE 2018, 13, e0208207. [Google Scholar]
  49. Goh, S.K.; Do, H.; Testro, A.; Pavlovic, J.; Vago, A.; Lokan, J.; Jones, R.M.; Christophi, C.; Dobrovic, A.; Muralidharan, V. The measurement of donor-specific cell-free DNA identifies recipients with biopsy-proven acute rejection requiring treatment after liver transplantation. Transplant. Direct. 2019, 5, e462. [Google Scholar] [CrossRef]
  50. Khush, K.K.; Patel, J.; Pinney, S.; Kao, A.; Alharethi, R.; DePasquale, E.; Ewald, G.; Berman, P.; Kanwar, M.; Hiller, D.; et al. Noninvasive detection of graft injury after heart transplant using donor-derived cell-free DNA: A prospective multicenter study. Am. J. Transplant. 2019, 19, 2889–2899. [Google Scholar] [CrossRef] [Green Version]
  51. Altuğ, Y.; Liang, N.; Ram, R.; Ravi, H.; Ahmed, E.; Brevnov, M.; Swenerton, R.K.; Zimmermann, B.; Malhotra, M.; Demko, Z.P.; et al. Analytical validation of a single-nucleotide polymorphism-based donor-derived cell-free DNA assay for detecting rejection in kidney transplant patients. Transplantation 2019, 103, 2657. [Google Scholar] [CrossRef] [Green Version]
  52. Sigdel, T.K.; Archila, F.A.; Constantin, T.; Prins, S.A.; Liberto, J.; Damm, I.; Towfighi, P.; Navarro, S.; Kirkizlar, E.; Demko, Z.P.; et al. Optimizing detection of kidney transplant injury by assessment of donor-derived cell-free DNA via massively multiplex PCR. J. Clin. Med. 2019, 8, 19. [Google Scholar] [CrossRef] [Green Version]
  53. Oellerich, M.; Shipkova, M.; Asendorf, T.; Walson, P.D.; Schauerte, V.; Mettenmeyer, N.; Kabakchiev, M.; Hasche, G.; Gröne, H.J.; Friede, T.; et al. Absolute quantification of donor-derived cell-free DNA as a marker of rejection and graft injury in kidney transplantation: Results from a prospective observational study. Am. J. Transplant. 2019, 19, 3087–3099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Sayah, D.; Weigt, S.S.; Ramsey, A.; Ardehali, A.; Golden, J.; Ross, D.J. Plasma Donor-derived Cell-free DNA Levels Are Increased During Acute Cellular Rejection After Lung Transplant: Pilot Data. Transplant. Direct. 2020, 6, e608. [Google Scholar] [CrossRef]
  55. Potter, S.R.; Hinojosa, R.; Miles, C.D.; O’brien, D.; Ross, D.J. Remote monitoring using donor-derived, cell-free DNA after kidney transplantation during the coronavirus disease 2019 pandemic. Kidney Res. Clin. Pract. 2020, 39, 495. [Google Scholar] [CrossRef]
  56. Schütz, E.; Asendorf, T.; Beck, J.; Schauerte, V.; Mettenmeyer, N.; Shipkova, M.; Wieland, E.; Kabakchiev, M.; Walson, P.D.; Schwenger, V.; et al. Time-dependent apparent increase in dd-cfDNA percentage in clinically stable patients between one and five years following kidney transplantation. Clin. Chem. 2020, 66, 1290–1299. [Google Scholar] [CrossRef]
  57. Shen, J.; Guo, L.; Yan, P.; Zhou, J.; Zhou, Q.; Lei, W.; Liu, H.; Liu, G.; Lv, J.; Liu, F.; et al. Prognostic value of the donor-derived cell-free DNA assay in acute renal rejection therapy: A prospective cohort study. Clin. Transplant. 2020, 34, e14053. [Google Scholar] [CrossRef]
  58. Puliyanda, D.P.; Swinford, R.; Pizzo, H.; Garrison, J.; De Golovine, A.M.; Jordan, S.C. Donor-derived cell-free DNA (dd-cfDNA) for detection of allograft rejection in pediatric kidney transplants. Pediatric Transplant. 2020, 25, e13850. [Google Scholar]
  59. Stites, E.; Kumar, D.; Olaitan, O.; John Swanson, S.; Leca, N.; Weir, M.; Bromberg, J.; Melancon, J.; Agha, I.; Fattah, H.; et al. High levels of dd-cfDNA identify patients with TCMR 1A and borderline allograft rejection at increased risk of graft injury. Am. J. Transplant. 2020, 20, 2491–2498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Khush, K.K.; De Vlaminck, I.; Luikart, H.; Ross, D.J.; Nicolls, M.R. Donor-derived, cell-free DNA levels by next-generation targeted sequencing are increased in allograft rejection after lung transplantation. ERJ Open Res. 2021, 7, 00462-2020. [Google Scholar] [CrossRef] [PubMed]
  61. Thongprayoon, C.; Vaitla, P.; Craici, I.M.; Thongprayoon, C.; Vaitla, P.; Craici, I.M.; Leeaphorn, N.; Hansrivijit, P.; Salim, S.A.; Bathini, T.; et al. The use of donor-derived cell-free DNA for assessment of allograft rejection and injury status. J Clin. Med. 2020, 9, 1480. [Google Scholar] [CrossRef]
  62. Bronkhorst, A.J.; Ungerer, V.; Holdenrieder, S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomol. Detect. Quantif. 2019, 17, 100087. [Google Scholar] [CrossRef]
  63. Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990. [Google Scholar] [CrossRef]
  64. Alekseeva, L.A.; Mironova, N.L.; Brenner, E.V.; Kurilshikov, A.M.; Patutina, O.A.; Zenkova, M.A. Alteration of the exDNA profile in blood serum of LLC-bearing mice under the decrease of tumour invasion potential by bovine pancreatic DNase I treatment. PLoS ONE 2017, 12, e0171988. [Google Scholar]
  65. Yu, S.C.; Lee, S.W.; Jiang, P.; Leung, T.Y.; Chan, K.A.; Chiu, R.W.; Lo, Y.D. High-resolution profiling of fetal DNA clearance from maternal plasma by massively parallel sequencing. Clin. Chem. 2013, 59, 1228–1237. [Google Scholar] [CrossRef] [Green Version]
  66. Gounden, V.; Bhatt, H.; Jialal, I. Renal function tests. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2018. [Google Scholar]
  67. Wilkinson, A. Protocol transplant biopsies: Are they really needed? Clin. J. Am. Soc. Nephrol. 2006, 1, 130–137. [Google Scholar] [CrossRef] [Green Version]
  68. Berl, T. American Society of Nephrology renal research report. J. Am. Soc. Nephrol. 2005, 16, 1886–1903. [Google Scholar]
  69. Rush, D.; Arlen, D.; Boucher, A.; Busque, S.; Cockfield, S.M.; Girardin, C.; Knoll, G.; Lachance, J.G.; Landsberg, D.; Shapiro, J.; et al. Lack of benefit of early protocol biopsies in renal transplant patients receiving TAC and MMF: A randomized study. Am. J. Transplant. 2007, 7, 2538–2545. [Google Scholar] [CrossRef] [PubMed]
  70. Gadi, V.K.; Nelson, J.L.; Boespflug, N.D.; Guthrie, K.A.; Kuhr, C.S. Soluble donor DNA concentrations in recipient serum correlate with pancreas-kidney rejection. Clin. Chem. 2006, 52, 379–382. [Google Scholar] [CrossRef] [Green Version]
  71. Mehta, S.G.; Chang, J.H.; Alhamad, T.; Bromberg, J.S.; Hiller, D.J.; Grskovic, M.; Yee, J.P.; Mannon, R.B. Repeat kidney transplant recipients with active rejection have increased donor-derived cell-free DNA. Am. J. Transplant. 2019, 19, 1597. [Google Scholar] [CrossRef]
  72. Gordon, P.M.; Khan, A.; Sajid, U.; Chang, N.; Suresh, V.; Dimnik, L.; Lamont, R.E.; Parboosingh, J.S.; Martin, S.R.; Pon, R.T.; et al. An algorithm measuring donor cell-free DNA in plasma of cellular and solid organ transplant recipients that does not require donor or recipient genotyping. Front. Cardiovasc. Med. 2016, 22, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Levine, D.J.; Ross, D.J.; Sako, E. Single Center “Snapshot” Experience With Donor-Derived Cell-Free DNA after Lung Transplantation. Biomark. Insights 2020, 15, 1177271920958704. [Google Scholar] [CrossRef]
  74. Richmond, M.E.; Zangwill, S.D.; Kindel, S.J.; Deshpande, S.R.; Schroder, J.N.; Bichell, D.P.; Knecht, K.R.; Mahle, W.T.; Wigger, M.A.; Gaglianello, N.A.; et al. Donor fraction cell-free DNA and rejection in adult and pediatric heart transplantation. J. Heart Lung Transplant. 2020, 39, 454–463. [Google Scholar] [CrossRef] [Green Version]
  75. Zhao, D.; Zhou, T.; Luo, Y.; Wu, C.; Xu, D.; Zhong, C.; Cong, W.; Liu, Q.; Zhang, J.; Xia, Q. Preliminary clinical experience applying donor-derived cell-free DNA to discern rejection in pediatric liver transplant recipients. Sci. Rep. 2021, 11, 1–9. [Google Scholar]
  76. Rinaldi, M.; Bartoletti, M.; Bussini, L.; Pancaldi, L.; Pascale, R.; Comai, G.; Morelli, M.; Ravaioli, M.; Cescon, M.; Cristini, F.; et al. COVID-19 in solid organ transplant recipients: No difference in survival compared to general population. Transpl. Infect. Dis. 2021, 23, 13421. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram of included articles.
Figure 1. PRISMA flow diagram of included articles.
Transplantology 02 00034 g001
Figure 2. Life cycle of donor-derived cell-free DNA, its identification and quantification. Abbreviations: dd-cfDNA, donor-derived cell-free DNA; SNP, single nucleotide polymorphism.
Figure 2. Life cycle of donor-derived cell-free DNA, its identification and quantification. Abbreviations: dd-cfDNA, donor-derived cell-free DNA; SNP, single nucleotide polymorphism.
Transplantology 02 00034 g002
Figure 3. Progression of organ transplant to graft failure. Elevation of dd-cfDNA can be an early indicator of graft damage, rejection and failure.
Figure 3. Progression of organ transplant to graft failure. Elevation of dd-cfDNA can be an early indicator of graft damage, rejection and failure.
Transplantology 02 00034 g003
Table 1. A timeline comparison of methods and techniques to assess dd-cfDNA to diagnose rejection.
Table 1. A timeline comparison of methods and techniques to assess dd-cfDNA to diagnose rejection.
NameOrgan(s)Sample Typedd-cfDNA MethodTechniqueDonor Sample Required
Zhang 1999 [26]KidneyUrineSex mismatch (SRY gene)Real-time quantitative PCRNo
Moreira 2009 [27]KidneyPlasma, UrineHemoglobin, beta (HBB) gene & TSPY1 geneQuantitative PCRYes
Synder 2011 [30]HeartPlasmaSex mismatch (Chr Y) & shotgun sequencingDigital PCRYes
Sigdel 2013 [29]KidneyUrineSex mismatch (SRY gene)Digital PCRNo
Beck 2013 [32]Heart, Kidney, LiverPlasmaInformative SNP panelDroplet digital PCRNo
Vlaminck 2014 [36]HeartPlasmaWhole-genome arraysQuantitative PCRYes
Macher 2014 [28]LiverPlasmaSRY & beta-globin geneReal time quantitative PCRNo
Vlaminck 2015 [37]LungsPlasmaWhole-genome arraysQuantitative PCRYes
Beck 2015 [38]Heart, KidneyPlasmaInformative SNP panelDroplet digital PCRNo
Burnham 2016 [39]LungsPlasmaSNP sequencing with mitochondrial reference sequencesQuantitative PCRYes
Macher 2016 [40]LiverPlasmaRH-negative recipient & RH-positive donor pairReal time PCRYes
Grskovic 2016 [33]Heart, KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Schutz 2016 [41]LiverPlasmaInformative SNP panelDroplet digital PCRNo
Zou 2017 [42]LungPlasmaHLA specific primers and probesDroplet digital PCRNo
Bromberg 2017 [43]KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Goh 2017 [44]LiverPlasmaDeletion/Insertion Polymorphism (DIP) panelDroplet digital PCRYes
Schutz 2017 [3]LiverPlasmaInformative SNP panelDroplet digital PCRNo
Bloom 2017 [45]KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Tanaka 2018 [46]LungPlasmaSNP genotyping & informative SNP panelDroplet digital PCRYes
Jordan 2018 [47]KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Weir 2018 [34]KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Beck 2018 [2]HeartPlasmaInformative SNP panelDroplet digital PCRNo
Gielis 2018 [48]KidneyPlasmaMultiplex PCR assayNext-generation sequencingYes
Goh 2019 [49]LiverPlasmaDeletion/Insertion Polymorphism (DIP) panelDroplet digital PCRYes
Khush 2019 [50]HeartPlasmaInformative SNP panelNext-generation sequencingNo
Altug 2019 [51]KidneyPlasmaBi-allelic SNP on Chr-2,13,18,21Massively multiplexed PCRNo
Sigdel 2019 [52]KidneyPlasmaSNP-based assay & LabChip NGS 5kMassively multiplex PCR-next-generation sequencingNo
Oellerich 2019 [53]KidneyPlasmaInformative SNP panelDroplet digital PCRNo
Sayah 2020 [54]LungPlasma Informative SNP panelNext-generation sequencingNo
Potter 2020 [55]KidneyPlasma Informative SNP panelNext-generation sequencingNo
Schutz 2020 [56]KidneyPlasma Informative SNP panelDroplet digital PCRNo
Shen 2020 [57]KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Puliyanda 2020 [58]Lung PlasmaInformative SNP panelNext-generation sequencingNo
Stites 2020 [59]KidneyPlasmaInformative SNP panelNext-generation sequencingNo
Khush 2021 [60]Lung PlasmaInformative SNP panelNext-generation sequencingNo
Abbreviations: Chr, chromosome; HLA, human leukocyte antigen; PCR, polymerase chain reaction; RH, Rhesus factor; SNP, Single nucleotide polymorphism; SRY, sex-determining region Y.
Table 2. Sensitivity and specificity of dd-cfDNA to detect graft rejection and/or damage.
Table 2. Sensitivity and specificity of dd-cfDNA to detect graft rejection and/or damage.
NameOrgan(s)Sample TypeSensitivity (%)Specificity (%)PPV (%)NPV (%)
Moiera 2009 [27]KidneyPlasma, Urine89855398
Sigdel 2013 [29]KidneyUrine8175--
Schutz 2016 [41]LiverPlasma91.292.5--
Schutz 2017 [3]LiverPlasma90.392.9--
Bloom 2017 [45]KidneyPlasma81834496
Jordan 2018 [47]KidneyPlasma81828183
Weir 2018 [34]KidneyPlasma69.68460.988.6
Khush 2019 [50]HeartPlasma80448.797.1
Sigdel 2019 [52]KidneyPlasma88.772.65295
Oellerich 2019 [53]KidneyPlasma73731398
Sayah 2020 [54]LungPlasma 73.152.93485.5
Levine 2020 [73]LungPlasma 81100--
Richmond 2020 [74]Heart Plasma 7579--
Puliyanda 2020 [58]Lung Plasma86100--
Zhao 2021 [75]Liver Plasma 81.881.956.293.9
Khush 2021 [60]Lung Plasma55.675.843.383.6
PPV: Positive predictive value; NPV: Negative predictive value. Mean sensitivities and specificities for dd-cfDNA to diagnose rejection are 79.83 ± 9.15% and 79.61 ± 14.96%, respectively.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kumar, N.; Rana, R.; Rana, D.S.; Gupta, A.; Sachdeva, M.P. Donor-Derived Cell-Free DNA to Diagnose Graft Rejection Post-Transplant: Past, Present and Future. Transplantology 2021, 2, 348-361. https://doi.org/10.3390/transplantology2030034

AMA Style

Kumar N, Rana R, Rana DS, Gupta A, Sachdeva MP. Donor-Derived Cell-Free DNA to Diagnose Graft Rejection Post-Transplant: Past, Present and Future. Transplantology. 2021; 2(3):348-361. https://doi.org/10.3390/transplantology2030034

Chicago/Turabian Style

Kumar, Naveen, Rashmi Rana, Devender Singh Rana, Anurag Gupta, and Mohinder Pal Sachdeva. 2021. "Donor-Derived Cell-Free DNA to Diagnose Graft Rejection Post-Transplant: Past, Present and Future" Transplantology 2, no. 3: 348-361. https://doi.org/10.3390/transplantology2030034

Article Metrics

Back to TopTop