cfDNA Analysis: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 1 by Isabel Legaz.

Degraded DNA fragments released into the blood or other fluids are known as cfDNA. Its first detection dates back to 1948 in patients with systemic lupus erythematosus.

  • donor-derived cell-free DNA (cfDNA)
  • graft injury
  • acute rejection
  • organ transplant

1. Introduction

In kidney transplantation, a biopsy is currently the gold standard for monitoring the transplanted organ. A biopsy involves taking a small tissue sample from the transplanted kidney, which is then examined under a microscope to assess organ health. This is done to assess for any signs of rejection or other problems that may be occurring in the transplanted kidney. However, this is far from an ideal screening method given its invasive nature and the discomfort it can cause the patient. Large-scale studies in renal transplantation show that approximately 1% of biopsies generate major complications, with a risk of macroscopic hematuria greater than 3.5% [1]. Most biopsy-related complications, such as pain and bleeding, are minor and localized, and can be managed conservatively [2]. The most severe complication, however, is the risk of perforation of the collecting system or the kidney itself, which can result in severe hemorrhage, sepsis, and even death. To minimize the risk of complications, careful patient selection, proper imaging guidance, and specialized instruments and techniques are essential [3,4][3][4]. Imaging techniques, such as ultrasound and computed tomography (CT), are essential for accurate needle placement. Ultrasound imaging is the most commonly used modality for needle guidance due to its versatility, cost-effectiveness, and relative safety. It allows for real-time visualization of the renal transplant and the surrounding anatomy, making it ideal for guiding percutaneous needle placement. CT imaging can also be used but is typically reserved for more complex cases with insufficient ultrasound imaging.
In addition, with current immunosuppressive therapies, the detection of subclinical rejection is too infrequent to justify this risk, which has meant that many units no longer perform these routine biopsies [5], thus raising the urgent need to find a new non-invasive biomarker that allows the detection of said rejection in order to intervene in time or modify immunosuppression. In response to this need, recent studies have shown that non-invasive biomarkers could be a viable option for detecting subclinical rejection [6,7,8,9][6][7][8][9]. These non-invasive biomarkers include urinary and serum markers such as urinary albumin-to-creatinine ratio (UACR) and donor-specific antibodies (DSA). Additionally, imaging techniques, such as magnetic resonance imaging (MRI) and ultrasound (US), have been used to detect graft changes that may signal rejection [10,11,12][10][11][12]. Finally, genetic and epigenetic biomarkers, such as microRNAs, have also been used to detect subclinical rejection [13,14,15][13][14][15]. Ultimately, using these non-invasive biomarkers could help identify and intervene in cases of subclinical rejection earlier, thus avoiding more serious complications.

2. Types de Cell-Free DNA

Degraded DNA fragments released into the blood or other fluids are known as cfDNA. Its first detection dates back to 1948 in patients with systemic lupus erythematosus [20][16]. It would not be until 1970 that this new biomarker would begin to be considered helpful for the clinic, as researchers observed differences in its concentration depending on the health status of the individual studied and began to see its application in cancer patients by allowing the detection of fragments of tumor DNA in the blood [21][17]. Its interest increased when it was discovered that tumor cells not only released cfDNA into the bloodstream but that these fragments also had the genetic and epigenetic changes of the tumor cells from which they had originated [22][18]. Shortly after, analysis of fetal cfDNA in maternal plasma began to be used to detect Rh mismatches and chromosomal aneuploidies [23][19]. Recent literature shows that different cfDNA types can be used as biomarkers of various disease states [16,24][20][21]. The following stand out for their relevance: ccf mtDNA (circulating cell-free mitochondrial DNA), ctDNA (circulating tumor DNA), cffDNA (cell-free fetal DNA), and dd-cfDNA (donor-derived cell-free DNA). These types and their applications are listed in Table 1.
Table 1.
Different types of cfDNA and their main clinical applications.

References

  1. Schwarz, A.; Gwinner, W.; Hiss, M.; Radermacher, J.; Mengel, M.; Haller, H. Safety and Adequacy of Renal Transplant Protocol Biopsies. Am. J. Transplant. 2005, 5, 1992–1996.
  2. Volpe, A.; Kachura, J.R.; Geddie, W.R.; Evans, A.J.; Gharajeh, A.; Saravanan, A.; Jewett, M.A.S. Techniques, Safety and Accuracy of Sampling of Renal Tumors by Fine Needle Aspiration and Core Biopsy. J. Urol. 2007, 178, 379–386.
  3. Kim, K.R.; Thomas, S. Complications of image-guided thermal ablation of liver and kidney neoplasms. Semin. Intervent. Radiol. 2014, 31, 138–148.
  4. Michel, M.S.; Trojan, L.; Rassweiler, J.J. Complications in Percutaneous Nephrolithotomy. Eur. Urol. 2007, 51, 899–906.
  5. Rush, D.N.; Gibson, I.W. Subclinical inflammation in renal transplantation. Transplantation 2019, 103, E139–E145.
  6. Mac, Q.D.; Mathews, D.V.; Kahla, J.A.; Stoffers, C.M.; Delmas, O.M.; Holt, B.A.; Adams, A.B.; Kwong, G.A. Non-invasive early detection of acute transplant rejection via nanosensors of granzyme B activity. Nat. Biomed. Eng. 2019, 3, 281–291.
  7. Hirt-Minkowski, P.; De Serres, S.A.; Ho, J. Developing renal allograft surveillance strategies-urinary biomarkers of cellular rejection. Can. J. Kidney Health Dis. 2015, 2, 28.
  8. Jamshaid, F.; Froghi, S.; Di Cocco, P.; Dor, F.J.M.F. Novel non-invasive biomarkers diagnostic of acute rejection in renal transplant recipients: A systematic review. Int. J. Clin. Pract. 2018, 72, e13220.
  9. Baumann, A.K.; Beck, J.; Kirchner, T.; Hartleben, B.; Schütz, E.; Oellerich, M.; Wedemeyer, H.; Jaeckel, E.; Taubert, R. Elevated fractional donor-derived cell-free DNA during subclinical graft injury after liver transplantation. Liver Transplant. 2022, 28, 1911–1919.
  10. Neimatallah, M.A.; Dong, Q.; Schoenberg, S.O.; Cho, K.J.; Prince, M.R. Magnetic Resonance Imaging in Renal Transplantation. J. Magn. Reson. Imaging Off. J. Int. Soc. Magn. Reson. Med. 1999, 10, 357–368.
  11. Hollis, E.; Shehata, M.; Khalifa, F.; Abou El-Ghar, M.; El-Diasty, T.; El-Baz, A. Towards non-invasive diagnostic techniques for early detection of acute renal transplant rejection: A review. Egypt. J. Radiol. Nucl. Med. 2017, 48, 257–269.
  12. Farag, A.; El-Baz, A.; Yuksel, S.E.; El-Ghar, M.A.; Eldiasty, T. A framework for the detection of acute renal rejection with dynamic contrast enhanced magnetic resonance imaging. In Proceedings of the 3rd IEEE International Symposium on Biomedical Imaging: Nano to Macro, Arlington, VA, USA, 6–9 April 2006; 2006, pp. 418–421.
  13. Bontha, S.V.; Maluf, D.G.; Mueller, T.F.; Mas, V.R. Systems Biology in Kidney Transplantation: The Application of Multi-Omics to a Complex Model. Am. J. Transplant. 2017, 17, 11–21.
  14. Asvapromtada, S.; Sonoda, H.; Kinouchi, M.; Oshikawa, S.; Takahashi, S.; Hoshino, Y.; Sinlapadeelerdkul, T.; Yokota-Ikeda, N.; Matsuzaki, T.; Ikeda, M. Recent Advances on Biomarkers of Early and Late Kidney Graft Dysfunction. Int. J. Mol. Sci. 2020, 21, 5404.
  15. Salvadori, M.; Tsalouchos, A. Biomarkers in renal transplantation: An updated review. World J. Transplant. 2017, 7, 161.
  16. Tug, S.; Helmig, S.; Menke, J.; Zahn, D.; Kubiak, T.; Schwarting, A.; Simon, P. Correlation between cell free DNA levels and medical evaluation of disease progression in systemic lupus erythematosus patients. Cell Immunol. 2014, 292, 32–39.
  17. Ulrich, B.C.; Guibert, N. Towards a comprehensive framework for cell-free DNA analysis: Lessons from TRACERx. Ann. Transl. Med. 2017, 5, 446–451.
  18. Arneth, B. Update on the types and usage of liquid biopsies in the clinical setting: A systematic review. BMC Cancer 2018, 18, 527.
  19. Jeon, S.H.; Lee, H.J.; Bae, K.; Yoon, K.A.; Lee, E.S.; Cho, Y. Efficient Capture and Isolation of Tumor-Related Circulating Cell-Free DNA from Cancer Patients Using Electroactive Conducting Polymer Nanowire Platforms. Theranostics 2016, 6, 828.
  20. Jimenez-Coll, V.; Llorente, S.; Boix, F.; Alfaro, R.; Galián, J.A.; Martinez-Banaclocha, H.; Botella, C.; Moya-Quiles, M.R.; Muro-Pérez, M.; Minguela, A.; et al. Monitoring of Serological, Cellular and Genomic Biomarkers in Transplantation, Computational Prediction Models and Role of Cell-Free DNA in Transplant Outcome. Int. J. Mol. Sci. 2023, 24, 3908.
  21. Pascual, J.; Attard, G.; Bidard, F.C.; Curigliano, G.; De Mattos-Arruda, L.; Diehn, M.; Italiano, A.; Lindberg, J.; Merker, J.D.; Montagut, C.; et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: A report from the ESMO Precision Medicine Working Group. Ann. Oncol. 2022, 33, 750–768.
  22. 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.
  23. Starzl, T.E.; Murase, N.; Ildstad, S.; Ricordi, C.; Demetris, A.J.; Trucco, M. Cell migration, chimerism, and graft acceptance. Lancet 1992, 339, 1579.
  24. 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.
  25. Lo, Y.M.D.; Tein, M.S.C.; Pang, C.C.P.; Yeung, C.K.; Tong, K.L.; Magnus Hjelm, N. Presence of donor-specific DNA in plasma of kidney and liver-transplant recipients. Lancet 1998, 351, 1329–1330.
  26. Kueht, M.L.; Dongur, L.P.; Cusick, M.; Stevenson, H.L.; Mujtaba, M. The Current State of Donor-Derived Cell-Free DNA Use in Allograft Monitoring in Kidney Transplantation. J. Pers. Med. 2022, 12, 1700.
  27. 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.
  28. 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.
  29. Sharon, E.; Shi, H.; Kharbanda, S.; Koh, W.; Martin, L.R.; Khush, K.K.; Valantine, H.; Pritchard, J.K.; De Vlaminck, I. Quantification of transplant-derived circulating cell-free DNA in absence of a donor genotype. PLoS Comput. Biol. 2017, 13, e1005629.
  30. 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.
  31. Huang, E.; Jordan, S.C. Donor-derived cell-free DNA in kidney transplantation: Evolving concepts and potential limitations. Kidney Int. 2022, 101, 676–677.
  32. Huang, E.; Sethi, S.; Peng, A.; Najjar, R.; Mirocha, J.; Haas, M.; Vo, A.; Jordan, S.C. Early clinical experience using donor-derived cell-free DNA to detect rejection in kidney transplant recipients. Am. J. Transplant. 2019, 19, 1663–1670.
  33. Clausen, F.B.; Jørgensen, K.M.C.L.; Wardil, L.W.; Nielsen, L.K.; Krog, G.R. Droplet digital PCR-based testing for donor-derived cell-free DNA in transplanted patients as noninvasive marker of allograft health: Methodological aspects. PLoS ONE 2023, 18, e0282332.
  34. 30876—Viracor TRAC® Kidney dd-cfDNA | Clinical | Eurofins-Viracor. Available online: https://www.eurofins-viracor.com/clinical/test-menu/30876-viracor-trac-kidney-dd-cfdna/ (accessed on 17 February 2023).
  35. 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. 2018, 8, 19.
  36. Halloran, P.F.; Reeve, J.; Madill-Thomsen, K.S.; Demko, Z.; Prewett, A.; Billings, P. The Trifecta Study: Comparing Plasma Levels of Donor-derived Cell-Free DNA with the Molecular Phenotype of Kidney Transplant Biopsies. J. Am. Soc. Nephrol. 2022, 33, 387–400.
  37. Muro, M. ¿Es el ADN libre circulante (cfDNA) derivado del donante (dd-cfDNA) un nuevo biomarcador para el rechazo de aloinjertos en trasplante? Inmunología 2020, 39, 23–26.
  38. Oellerich, M.; Sherwood, K.; Keown, P.; Schütz, E.; Beck, J.; Stegbauer, J.; Rump, L.C.; Walson, P.D. Liquid biopsies: Donor-derived cell-free DNA for the detection of kidney allograft injury. Nat. Rev. Nephrol. 2021, 17, 591–603.
  39. Puttarajappa, C.M.; Mehta, R.B.; Roberts, M.S.; Smith, K.J.; Hariharan, S. Economic analysis of screening for subclinical rejection in kidney transplantation using protocol biopsies and noninvasive biomarkers. Am. J. Transplant. 2021, 21, 186–197.
  40. Porrini, E.; Ruggenenti, P.; Luis-Lima, S.; Carrara, F.; Jiménez, A.; de Vries, A.P.J.; Torres, A.; Gaspari, F.; Remuzzi, G. Estimated GFR: Time for a critical appraisal. Nat. Rev. Nephrol. 2018, 15, 177–190.
  41. Filippone, E.J.; Farber, J.L. The Monitoring of Donor-derived Cell-free DNA in Kidney Transplantation. Transplantation 2021, 105, 509–516.
  42. Kataria, A.; Kumar, D.; Gupta, G. Donor-derived Cell-free DNA in Solid-organ Transplant Diagnostics: Indications, Limitations, and Future Directions. Transplantation 2021, 105, 1203–1211.
  43. O’Callaghan, J.M.; Knight, S.R. Noninvasive biomarkers in monitoring kidney allograft health. Curr. Opin. Organ Transplant. 2019, 24, 411–415.
  44. Rosenheck, J.P.; Keller, B.C.; Fehringer, G.; Demko, Z.P.; Bohrade, S.M.; Ross, D.J. Why Cell-Free DNA Can Be a “Game Changer” for Lung Allograft Monitoring for Rejection and Infection. Curr. Pulmonol. Rep. 2022, 11, 75–85.
  45. Oellerich, M.; Budde, K.; Osmanodja, B.; Bornemann-Kolatzki, K.; Beck, J.; Schütz, E.; Walson, P.D. Donor-derived cell-free DNA as a diagnostic tool in transplantation. Front. Genet. 2022, 13, 1031894.
  46. 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.
  47. Whitlam, J.B.; Ling, L.; Skene, A.; Kanellis, J.; Ierino, F.L.; Slater, H.R.; Bruno, D.L.; Power, D.A. Diagnostic application of kidney allograft-derived absolute cell-free DNA levels during transplant dysfunction. Am. J. Transplant. 2019, 19, 1037–1049.
  48. Oellerich, M.; Wu, A.; Halloran, P.F.; De Vlaminck, I.; Keller, M.; Agbor-Enoh, S. Molecular Approaches to Transplant Monitoring; Is the Horizon Here? Clin. Chem. 2021, 67, 1443–1449.
  49. Oellerich, M.; Budde, K.; Osmanodja, B.; Bornemann-Kolatzki, K.; Beck, J.; Schütz, E.; Walson, P.D. Donor-Derived Cell-free DNA for Personalized Immunosuppression in Renal Transplantation. Ther. Drug Monit. 2023, 45, 20–25.
  50. Lum, E.L.; Nieves-Borrero, K.; Homkrailas, P.; Lee, S.; Danovitch, G.; Bunnapradist, S. Single center experience comparing two clinically available donor derived cell free DNA tests and review of literature. Transplant. Rep. 2021, 6, 100079.
  51. Goussous, N.; Xie, W.; Dawany, N.; Scalea, J.R.; Bartosic, A.; Haririan, A.; Kalil, R.; Drachenberg, C.; Costa, N.; Weir, M.R.; et al. Donor-derived Cell-free DNA in Infections in Kidney Transplant Recipients: Case Series. Transplant. Direct 2020, 6, e568.
  52. Kant, S.; Bromberg, J.; Haas, M.; Brennan, D. Donor-derived Cell-free DNA and the Prediction of BK Virus-associated Nephropathy. Transplant. Direct 2020, 6, e622.
More
Video Production Service