Next Article in Journal
Sarcopenic Obesity in Cervical Carcinoma: A Strong and Independent Prognostic Factor beyond the Conventional Predictors (ESTHER Study—AFRAID Project)
Previous Article in Journal
Blood Clot Dynamics and Fibrinolysis Impairment in Cancer: The Role of Plasma Histones and DNA
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Circulating Tumor DNA Profiling in Liver Transplant for Hepatocellular Carcinoma, Cholangiocarcinoma, and Colorectal Liver Metastases: A Programmatic Proof of Concept

by
Hanna Hong
1,†,
Chase J. Wehrle
1,*,†,
Mingyi Zhang
1,
Sami Fares
1,
Henry Stitzel
1,
David Garib
1,
Bassam Estfan
2,
Suneel Kamath
2,
Smitha Krishnamurthi
2,
Wen Wee Ma
2,
Teodora Kuzmanovic
2,
Elizabeth Azzato
3,
Emrullah Yilmaz
2,
Jamak Modaresi Esfeh
4,
Maureen Whitsett Linganna
4,
Mazhar Khalil
1,
Alejandro Pita
1,
Andrea Schlegel
1,
Jaekeun Kim
1,
R. Matthew Walsh
1,
Charles Miller
1,
Koji Hashimoto
1,
David Choon Hyuck Kwon
1 and
Federico Aucejo
1
add Show full author list remove Hide full author list
1
Department of Hepato-Pancreato-Biliary & Liver Transplant Surgery, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
2
Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
3
Molecular Pathology and Cytogenomics, Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
4
Department of Gastroenterology, Hepatology, and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2024, 16(5), 927; https://doi.org/10.3390/cancers16050927
Submission received: 26 January 2024 / Revised: 15 February 2024 / Accepted: 22 February 2024 / Published: 25 February 2024

Abstract

:

Simple Summary

Circulating tumor DNA (ctDNA) is emerging as a diagnostic and surveillance tool in cancer and recurrence. The recurrence rates after liver transplant for cancer are significant, highlighting the need for early detection and treatment. We report a cohort of patients who underwent liver transplant for hepatocellular carcinoma, cholangiocarcinoma, or colorectal cancer liver metastasis and received ctDNA testing pre- and/or post-transplant. We aim to show how ctDNA testing can be incorporated into pre-transplant work-up and post-transplant surveillance and discuss the benefits of this testing modality in the identification of genetic targets and surveillance of recurrence.

Abstract

Introduction: Circulating tumor DNA (ctDNA) is emerging as a promising, non-invasive diagnostic and surveillance biomarker in solid organ malignancy. However, its utility before and after liver transplant (LT) for patients with primary and secondary liver cancers is still underexplored. Methods: Patients undergoing LT for hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and colorectal liver metastases (CRLM) with ctDNA testing were included. CtDNA testing was conducted pre-transplant, post-transplant, or both (sequential) from 11/2019 to 09/2023 using Guardant360, Guardant Reveal, and Guardant360 CDx. Results: 21 patients with HCC (n = 9, 43%), CRLM (n = 8, 38%), CCA (n = 3, 14%), and mixed HCC/CCA (n = 1, 5%) were included in the study. The median follow-up time was 15 months (range: 1–124). The median time from pre-operative testing to surgery was 3 months (IQR: 1–4; range: 0–5), and from surgery to post-operative testing, it was 9 months (IQR: 2–22; range: 0.4–112). A total of 13 (62%) patients had pre-transplant testing, with 8 (62%) having ctDNA detected (ctDNA+) and 5 (32%) not having ctDNA detected (ctDNA-). A total of 18 (86%) patients had post-transplant testing, 11 (61%) of whom were ctDNA+ and 7 (33%) of whom were ctDNA-. The absolute recurrence rates were 50% (n = 5) in those who were ctDNA+ vs. 25% (n = 1) in those who were ctDNA- in the post-transplant setting, though this difference was not statistically significant (p = 0.367). Six (29%) patients (HCC = 3, CCA = 1, CRLM = 2) experienced recurrence with a median recurrence-free survival of 14 (IQR: 6–40) months. Four of these patients had positive post-transplant ctDNA collected following diagnosis of recurrence, while one patient had positive post-transplant ctDNA collected preceding recurrence. A total of 10 (48%) patients had sequential ctDNA testing, of whom n = 5 (50%) achieved ctDNA clearance (+/−). The remainder were ctDNA+/+ (n = 3, 30%), ctDNA−/− (n = 1, 10%), and ctDNA−/+ (n = 1, 11%). Three (30%) patients showed the acquisition of new genomic alterations following transplant, all without recurrence. Overall, the median tumor mutation burden (TMB) decreased from 1.23 mut/Mb pre-transplant to 0.00 mut/Mb post-transplant. Conclusions: Patients with ctDNA positivity experienced recurrence at a higher rate than the ctDNA- patients, indicating the potential role of ctDNA in predicting recurrence after curative-intent transplant. Based on sequential testing, LT has the potential to clear ctDNA, demonstrating the capability of LT in the treatment of systemic disease. Transplant providers should be aware of the potential of donor-derived cell-free DNA and improved approaches are necessary to address such concerns.

1. Introduction

Liver transplant is the only curative-intent treatment option for patients with unresectable hepatocellular carcinoma, cholangiocarcinoma, and colorectal liver metastasis [1,2,3,4,5,6]. However, the recurrence rates may be as high as 20–50% in certain conditions, highlighting the need for thorough and frequent post-transplant monitoring [3,4,7,8,9]. Traditional surveillance relies on cross-sectional imaging and serum biomarkers such as alpha-fetoprotein (AFP), carbohydrate/cancer antigen 19-9 (CA19-9), or carcinoembryonic antigen (CEA) depending on the disease. However, post-transplant recurrence remains a challenge in terms of diagnostics and treatment due to the limited sensitivity and specificity of current tools for cancer detection [10,11,12,13,14].
To address this issue, ctDNA-based liquid biopsy has emerged as a non-invasive approach that allows for the real-time monitoring of tumor dynamics, detection of minimal residual disease, and identification of actionable mutations [15,16,17,18,19]. In patients undergoing liver transplant, the use of cell-free DNA has also been applied to detecting rejection [20]. We have previously reported the use of ctDNA in patients undergoing liver transplant for CRLM [21]. However, its use as a predictive tool of recurrence in liver transplant remains to be fully explored.
Herein, we present a cohort of patients who underwent liver transplant for HCC, CCA, or CRLM and ctDNA testing at pre-transplant and/or post-transplant time points, demonstrating a proof of concept for ctDNA in this setting.

2. Methods

Patients who underwent liver transplantation for CRLM, HCC, or CCA with pre-transplant and/or post-transplant ctDNA assessment between November 2019 and September 2023 at a single quaternary care academic institution were included in this study. All the patients were evaluated by a multidisciplinary liver tumor board and liver transplant review committee. The demographic and clinical variables, including on imaging, laboratory values, and treatment courses, were collected via a retrospective review of the patients’ health charts, as approved by the Institutional Review Board (IRB).
The ctDNA was assessed using Guardant360, Guardant360 CDx, and Guardant Reveal assays (Guardant Health, Redwood City, CA, USA). Guardant360 uses next-generation sequencing (NGS) to detect clinically relevant genomic alterations in the circulating tumor DNA in plasma collected via the peripheral blood. NGS testing was performed as part of the standard clinical care in a CLIA-certified and College of American Pathologists-accredited laboratory. The blood was collected in two to four 10 mL Streck tubes, and the processed plasma was evaluated for single-nucleotide variants (SNVs), insertions–deletions (indels), gene fusions/rearrangements, and copy number variants (CNVs) across 83 genes. Mutations were annotated using OncoKB to define pathogenic variants. The blood tumor mutational burden (bTMB) was determined by analyzing the somatic SNVs and indels across a 1.0 Mb genomic backbone. For the TMB algorithm, common cancer drivers and resistance alterations, as well as putative CHIP alterations, were filtered from the analysis. Guardant Reveal uses NGS to determine the presence of ctDNA by assessing somatic alterations (SNVs, insertion–deletion alterations) and epigenomic signatures (methylation status). Guardant Reveal was used for the portion of patients with CRLM, while Guardant360 CDx was used for the portion of patients with HCC. Guardant360 was used for all cancer types.
Prior to January 2021, the ctDNA was collected and evaluated at the discretion of the treating surgeon. From January 2021 onward, attempts were made to collect the ctDNA at times outlined by the current institutional protocol of within 30 days pre-operatively, 30–60 days post-operatively, and every 3–6 months afterward (Figure 1). Synonymous mutations were excluded from the analysis.
Discrete variables were presented as frequency and percentages, and continuous variables were presented as medians with interquartile ranges due to non-normal distributions. Statistical analysis was performed using IBM SPSS Statistics Version 29.0 (Armonk, New York, NY, USA). A two-sided p-value < 0.05 was considered significant for all tests.

3. Results

A total of 21 patients underwent ctDNA testing and LT for HCC (n = 9, 43%), CRLM (n = 8, 38%), CCA (n = 3, 14%), and mixed HCC/CCA (n = 1, 5%) (Table 1). Nine (43%) patients underwent living donor liver transplant (LDLT), seven (33%) underwent orthotopic liver transplant (OLT) with grafts from donation after brain death (DBD), and five (24%) had OLT with grafts from donation after cardiac death (DCD) (Table 1). Most patients had cirrhosis (n = 19, 90%), with a median MELD score of 15 at the time of transplant. NASH (n = 6, 30%) was the most frequent cause of cirrhosis. The median tumor marker levels at the time of liver cancer diagnosis were AFP = 8 ng/mL (HCC), CA19-9 = 23 U/mL (CCA), and CEA = 31 ng/mL (CRLM). (Table 1). Prior to transplant, most patients (n = 18, 86%) received treatment, the most common being chemotherapy (n = 10, 48%), radiation (n = 6, 29%), TACE (n = 6, 29%), and TARE (n = 5, 24%). The post-transplant tumor marker levels were 3 ng/mL, 13 U/mL, and 1.8 ng/mL for AFP, CA19-9, and CEA, respectively (Table 1). Six (29%) patients (HCC = 3, CCA = 1, CRLM = 2) experienced recurrence with a median recurrence-free survival of 14 (IQR: 6-40) months. Two (10%) patients experienced cancer-related death, both with a diagnosis of HCC (Table 1). Overall, four (19%) patients experienced mortality, with a median overall survival of 16 (IQR: 8–40) months. The median and maximum follow-up times were 15 and 124 months, respectively (Table 1).
In terms of transplant, most patients (n = 18, 86%) underwent the piggyback technique (Table 2). The median warm ischemia time was 43 (IQR: 39–46) minutes, with a median LT duration of 589 (IQR: 471–702) minutes. Post-operatively, most patients underwent induction immunosuppression with basiliximab (n = 12, 57%) and initial immunosuppression with glucocorticoids, mycophenolate mofetil, and tacrolimus (n = 19, 90%) (Table 3). Five (24%) patients experienced bile leaks, requiring ERCP and/or PTHC, while three (14%) patients experienced biliary strictures requiring ERCP (Table 3). One patient had ischemic cholangiopathy and hepatic artery stenosis in addition to their biliary leak, requiring HJ reconstruction, PTHC, re-transplant, and stent placement (Table 3). Two patients experienced mild acute rejection, which was treated with IV steroids. No patients experienced chronic rejection (Table 3).
A total of 18 (86%) patients had post-transplant ctDNA, with 11 having ctDNA detected and 7 not having ctDNA detected (Table 4). The absolute recurrence rates were higher in patients with detected ctDNA (n = 5, 50%) compared to patients without ctDNA detected (n = 1, 25%), although this difference was not found to be statistically significant (p = 0.367).
Of the six (29%) patients with recurrence, five patients had post-transplant ctDNA detected. The remaining patient (#20) did not have post-transplant ctDNA detected during or after treatment of their recurrence with metastasectomy and chemotherapy (Table 4 and Table 5). Of the post-transplant ctDNA+ patients, 4/5 had ctDNA detected following radiologic detection of recurrence, while 1/5 (#7) had ctDNA detected prior to recurrence, without elevated tumor markers. Overall, only 3/6 patients (#3, 12, 21) had elevated serum tumor markers preceding recurrence, while 3/6 (#6, 7, 20) patients lacked elevation of the traditionally used tumor markers prior to recurrence.
Of all 21 patients, 10 (48%) patients had sequential ctDNA testing, with half (n = 5, 50%) having ctDNA clearance (+/−). The remainder were ctDNA+/+ (n = 3, 30%), ctDNA−/− (n = 1, 10%), and ctDNA−/+ (n = 1, 10%). More specifically, patients #9, 11, 15, 16, and 18 were ctDNA+/−; patients #2, 17, and 21 were ctDNA +/+; patient #13 was ctDNA−/−; and patient #14 was ctDNA−/+. Of note, patient #21 experienced recurrence. Three (30%) patients showed the acquisition of new genomic alterations in post-transplant ctDNA (#2, 14, 17) (Table 6). Patients #14 and 17 had no evidence of histopathologic viable tumors on explant (Table 7), suggesting potential alternate sources of these ctDNA findings. No signs of acute rejection were noticed for these patients (#14, #17, Table 6).
Overall, the median TMB decreased from 1.23 mut/Mb pre-transplant (n = 9) to 0.00 mut/Mb post-transplant (n = 11). For HCC, a TERT promoter mutation was the most common genomic alteration both pre-transplant and post-transplant (Figure 2). For CRLM, TP53 and APC mutations were the most common alterations observed pre-transplant, compared to NF1 and PTPN11 post-transplant (Figure 2).

4. Discussion

Liver transplant as a treatment for primary and secondary liver malignancy has grown in volume, with expansion from HCC to CCA and, more recently, to CRLM [6]. However, recurrence after LT remains a concern [22]. CtDNA has emerged as a non-invasive surveillance tool in predicting and detecting recurrence after the treatment of hepatic malignancies [23]. Compared to traditionally used tumor markers (e.g., CA19-9) which are notorious for their limited sensitivity and specificity, ctDNA offers a more individualized testing modality that can be used to predict recurrence-free survival at earlier time points, leading to guided decision-making for treatment selection [24,25].
This study demonstrates proof-of-concept for ctDNA testing in patients undergoing LT for primary and secondary liver cancers. We found a higher absolute recurrence rate in patients with positive post-transplant ctDNA. In patients who experienced recurrence, ctDNA was detected in all patients with active disease. Conversely, ctDNA was not detected in the one patient who achieved remission after recurrence. When comparing pre- vs. post-transplant ctDNA, clearance of ctDNA was observed in half of the patients who underwent sequential testing. An overall reduction in the TMB was also noted after LT. Interestingly, 30% of patients with sequential testing acquired new genomic alterations in post-transplant ctDNA, which may induce caution toward recurrent malignancy and/or the introduction of confounding genomic material that influences the interpretation of the results.
Our group previously published on the use of ctDNA in the context of hepatic resection for CRLM, showing how the detection of post-operative ctDNA was associated with an increased likelihood of disease recurrence [21]. Similarly, Tie et al. (2023) [24], Liu et al. (2023) [26], and Nishioka et al. (2022) [27] showed that post-operative ctDNA positivity predicts a reduced recurrence-free (RFS) and overall survival (OS) in patients undergoing hepatectomy for CRLM. The results of the GALAXY study further demonstrate the association of post-operative ctDNA with an increased recurrence risk and the ability to identify patients who derived benefits from adjuvant chemotherapy in patients with stage II or III CRC [28]. In patients with resected CCA, the preliminary results from Yoo at al. (2023) similarly show positive ctDNA status is predictive of a poor RFS [29]. In HCC, Wang et al. (2020) showed a reduced RFS with post-operative ctDNA assessed according to a panel of four hotspot genomic mutations in TP53 (G747T), CTNNB1 (A121G, C133T), and TERT (c.-124C>T) [30]. In the setting of liver transplant for unresectable primary liver cancer, larger scale studies by Huang et al. (2023) [23] and Jiang et al. (2022) [31] again display higher recurrence rates in patients with positive post-transplant ctDNA and decreased disease-free survival.
The widely known limitations of tumor serum biomarkers are additionally observed in our study. Of the six patients in our study who experienced recurrence, three (#6, 7, 20) had normal serum levels of traditionally used biomarkers at time of recurrence. However, ctDNA was detected post-transplant in two of these patients (#6, 7), demonstrating a potential set of patients in whom the recurrence of HCC following LT may be predicted or detected with ctDNA. To this end, expanding the enrollment of patients undergoing post-transplant ctDNA testing and conducting serial testing at earlier time points following LT may help elucidate whether the detection of ctDNA correlates with or predicts recurrence. If shown to be of prognostic utility, ctDNA could be used to stratify patients based on their risk of recurrence and determine more targeted, individualized selection of adjuvant therapy.
In addition, we report the acquisition of new mutations post-transplant in several patients who underwent sequential tumor-agnostic ctDNA testing. Although the exact source of the ctDNA is unknown, the absence of viable tumors in the explant histopathology for at least two patients may lead us to postulate that these mutations may be of donor origin. Alternatively, they may represent somatic mutations in the setting of immunosuppression post-transplant or clonal evolution. To address this concern, tumor-informed genetic testing may be considered due to its ability to differentiate ctDNA from germline-derived variants, clonal hematopoiesis of indeterminate potential, and dd-cfDNA. Such tumor-informed tools have been developed and are actively being explored in clinical studies and trials [32]. However, these methods do have limitations in patients who have received extensive pre-LT locoregional and systemic therapy, as adequate viable tumor is necessary for tissue-informed testing. Given the uncertain origin of the novel post-LT genomic alterations, making ctDNA-based treatment decisions may be challenging in this subset. At a minimum, pre- and post-LT testing should be pursued when using tissue-agnostic testing in order to obtain a pre-transplant comparison. With expanding evidence supporting the use of ctDNA testing in liver cancers [24,25,26,27,28,29,30,31], the optimization of protocols effective at addressing the concerns regarding donor-derived alterations is warranted in future studies.
In addition to assessing for the presence of ctDNA, liquid biopsy can identify specific genes that predict patient outcomes based on cancer. For example, in HCC, CTNNB1 and TERT have been shown to be two of the most commonly mutated genes and were present frequently in our cohort [33]. The presence of these two mutations, along with a mutation in TP53, in post-operative ctDNA has been associated with a decreased recurrence-free survival [30]. In CCA, the mutations are thought to be more heterogeneous, though mutations in KRAS, IDH1/2, FGFR, ERBB2, and BRAF have been noted to be more frequently mutated [34]. In colorectal cancer, mutations in APC and TP53 are known to drive the transition from adenoma to adenocarcinoma [35,36,37,38]. In patient #21, the presence of these mutations post-transplant, although at lower variant allele frequencies, was detected prior to diagnosis of recurrence (Table 8). While our study was not aimed at addressing the prognostic or therapeutic implications of specific genes, the correlation between our findings in solid organ transplant patients and the published findings in the non-transplant population is encouraging for the application of liquid biopsy to this new set of patients. Tissue-agnostic ctDNA testing could theoretically provide such analysis before transplant, allowing for pre-transplant prognostication. One example of potential utility is the detection of mutations that are contraindications to transplant, such as BRAF V600E, which represents a contraindication to LT for CRLM in our center. As detection of such a mutation pre-LT may preclude transplant due to high risk of recurrence, the use of ctDNA in the transplant population warrants further investigation for optimization of protocols and interpretation.
The limitations of this study include a small sample size, which is insufficient for determining causal relationships between ctDNA clearance and liver transplant. Furthermore, a low number of patients had sequential testing, which interferes with the evaluation of donor-derived cell-free DNA. Inconsistency in the ctDNA sampling and timing may have arisen due to challenges in clinical practice and logistics. To address these issues, a large-scale multi-institutional study is being conducted to increase the patient volume, and new institutional protocols have been implemented to ensure adequate sampling. Furthermore, the impact of neoadjuvant and adjuvant chemo, immune, and radiation therapy on the ctDNA results is still unknown. Lastly, the correlation of ctDNA with tissue-based mutational profiles was not assessed in the present study, although concurrent tissue testing is now ongoing.

5. Conclusions

Circulating tumor DNA can help us to identify recurrence after liver transplant for hepatic malignancy. Transplantation was also associated with clearance of the ctDNA burden in half of the patients with sequential testing. We report a subset of patients with non-viable tumors and novel post-transplant genomic profiles, raising concern about donor-derived sources; improved approaches are necessary to address the potential of such findings confounding treatment decisions. Larger-scale studies and serial monitoring should be conducted to confirm the utility of ctDNA as a surveillance tool for MRD post-transplant and optimize the timing of the screening protocols.

Author Contributions

Conceptualization, H.H., C.J.W. and F.A.; methodology, H.H. and C.J.W.; software, H.H. and C.J.W.; validation, H.H., C.J.W. and F.A.; formal analysis, H.H. and C.J.W.; investigation, H.H., M.Z., S.F., H.S. and D.G.; resources, F.A., D.C.H.K., K.H., C.M., R.M.W., J.K., A.S., A.P. and M.K.; data curation, H.H., C.J.W., F.A., M.Z., B.E., S.K. (Suneel Kamath), S.K. (Smitha Krishnamurthi), W.W.M., T.K., E.A., E.Y., J.M.E., M.W.L., M.K., A.P., A.S. and J.K.; writing—original draft preparation, H.H., C.J.W. and F.A.; writing—review and editing, all authors; visualization, all authors; supervision, F.A.; project administration, F.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

IRB approval was obtained under Cleveland Clinic’s IRB, 09-096.

Informed Consent Statement

Consent was waived due to the retrospective nature of the review.

Data Availability Statement

The data presented in the study are available in the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hagness, M.; Foss, A.; Line, P.-D.; Scholz, T.; Jørgensen, P.F.; Fosby, B.; Boberg, K.M.; Mathisen, O.; Gladhaug, I.P.; Egge, T.S.; et al. Liver Transplantation for Nonresectable Liver Metastases from Colorectal Cancer. Ann. Surg. 2013, 257, 800–806. [Google Scholar] [CrossRef] [PubMed]
  2. Dueland, S.; Guren, T.K.; Hagness, M.; Glimelius, B.; Line, P.-D.; Pfeiffer, P.; Foss, A.; Tveit, K.M. Chemotherapy or Liver Transplantation for Nonresectable Liver Metastases from Colorectal Cancer? Ann. Surg. 2015, 261, 956–960. [Google Scholar] [CrossRef]
  3. Dueland, S.; Syversveen, T.; Solheim, J.M.; Solberg, S.; Grut, H.; Bjørnbeth, B.A.; Hagness, M.; Line, P.-D. Survival Following Liver Transplantation for Patients with Nonresectable Liver-Only Colorectal Metastases. Ann. Surg. 2020, 271, 212–218. [Google Scholar] [CrossRef]
  4. Tabrizian, P.; Holzner, M.L.; Mehta, N.; Halazun, K.; Agopian, V.G.; Yao, F.; Busuttil, R.W.; Roberts, J.; Emond, J.C.; Samstein, B.; et al. Ten-Year Outcomes of Liver Transplant and Downstaging for Hepatocellular Carcinoma. JAMA Surg. 2022, 157, 779–788. [Google Scholar] [CrossRef]
  5. Twohig, P.; Peeraphatdit, T.B.; Mukherjee, S. Current Status of Liver Transplantation for Cholangiocarcinoma. World J. Gastrointest. Surg. 2022, 14, 1–11. [Google Scholar] [CrossRef]
  6. Gorji, L.; Brown, Z.J.; Limkemann, A.; Schenk, A.D.; Pawlik, T.M. Liver Transplant as a Treatment of Primary and Secondary Liver Neoplasms. JAMA Surg. 2024, 159, 211–218. [Google Scholar] [CrossRef]
  7. Agarwal, P.D.; Lucey, M.R. Management of Hepatocellular Carcinoma Recurrence after Liver Transplantation. Ann. Hepatol. 2022, 27, 100654. [Google Scholar] [CrossRef]
  8. Wu, T.C.; Smith, C.P.; Li, J.S.; Burton, J.; Jackson, N.J.; Tao, R.; Ludmir, E.B.; Raldow, A.C. A Systematic Review and Meta-Analysis of Pathologic Complete Response Rates for Patients with Cholangiocarcinoma Treated on Liver Transplant Protocols. J. Surg. Oncol. 2023, 129, 574–583. [Google Scholar] [CrossRef]
  9. Solheim, J.M.; Dueland, S.; Line, P.-D.; Hagness, M. Transplantation for Nonresectable Colorectal Liver Metastases: Long-Term Follow-Up of the First Prospective Pilot Study. Ann. Surg. 2023, 278, 239. [Google Scholar] [CrossRef] [PubMed]
  10. Berenguer, M.; Burra, P.; Ghobrial, M.; Hibi, T.; Metselaar, H.; Sapisochin, G.; Bhoori, S.; Kwan Man, N.; Mas, V.; Ohira, M.; et al. Posttransplant Management of Recipients Undergoing Liver Transplantation for Hepatocellular Carcinoma. Working Group Report From the ILTS Transplant Oncology Consensus Conference. Transplantation 2020, 104, 1143. [Google Scholar] [CrossRef] [PubMed]
  11. Hanif, H.; Ali, M.J.; Susheela, A.T.; Khan, I.W.; Luna-Cuadros, M.A.; Khan, M.M.; Lau, D.T.-Y. Update on the Applications and Limitations of Alpha-Fetoprotein for Hepatocellular Carcinoma. World J. Gastroenterol. 2022, 28, 216–229. [Google Scholar] [CrossRef]
  12. Lin, M.-S.; Huang, J.-X.; Yu, H. Elevated Serum Level of Carbohydrate Antigen 19-9 in Benign Biliary Stricture Diseases Can Reduce Its Value as a Tumor Marker. Int. J. Clin. Exp. Med. 2014, 7, 744–750. [Google Scholar] [PubMed]
  13. Roles of Serum and Biliary CEA, CA19-9, VEGFR3, and TAC in Differentiating between Malignant and Benign Biliary Obstructions. Available online: http://turkjgastroenterol.org/en/roles-of-serum-and-biliary-cea-ca19-9-vegfr3-and-tac-in-differentiating-between-malignant-and-benign-biliary-obstructions-134337 (accessed on 25 January 2024).
  14. Sekiguchi, M.; Matsuda, T. Limited Usefulness of Serum Carcinoembryonic Antigen and Carbohydrate Antigen 19-9 Levels for Gastrointestinal and Whole-Body Cancer Screening. Sci. Rep. 2020, 10, 18202. [Google Scholar] [CrossRef] [PubMed]
  15. Liquid Biopsy at the Frontier of Detection, Prognosis and Progression Monitoring in Colorectal Cancer—PMC. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8951719/ (accessed on 25 January 2024).
  16. Bent, A.; Raghavan, S.; Dasari, A.; Kopetz, S. The Future of ctDNA-Defined Minimal Residual Disease: Personalizing Adjuvant Therapy in Colorectal Cancer. Clin. Color. Cancer 2022, 21, 89–95. [Google Scholar] [CrossRef]
  17. Kopystecka, A.; Patryn, R.; Leśniewska, M.; Budzyńska, J.; Kozioł, I. The Use of ctDNA in the Diagnosis and Monitoring of Hepatocellular Carcinoma—Literature Review. Int. J. Mol. Sci. 2023, 24, 9342. [Google Scholar] [CrossRef]
  18. Cabel, L.; Proudhon, C.; Buecher, B.; Pierga, J.-Y.; Bidard, F.-C. Circulating Tumor DNA Detection in Hepatocellular Carcinoma. Ann. Oncol. 2018, 29, 1094–1096. [Google Scholar] [CrossRef]
  19. Li, Y.; Zheng, Y.; Wu, L.; Li, J.; Ji, J.; Yu, Q.; Dai, W.; Feng, J.; Wu, J.; Guo, C. Current Status of ctDNA in Precision Oncology for Hepatocellular Carcinoma. J. Exp. Clin. Cancer Res. 2021, 40, 140. [Google Scholar] [CrossRef]
  20. Levitsky, J.; Kandpal, M.; Guo, K.; Kleiboeker, S.; Sinha, R.; Abecassis, M. Donor-Derived Cell-Free DNA Levels Predict Graft Injury in Liver Transplant Recipients. Am. J. Transplant. 2022, 22, 532–540. [Google Scholar] [CrossRef]
  21. Wehrle, C.J.; Raj, R.; Aykun, N.; Orabi, D.; Estfan, B.; Kamath, S.; Krishnamurthi, S.; Fujiki, M.; Hashimoto, K.; Quintini, C.; et al. Liquid Biopsy by ctDNA in Liver Transplantation for Colorectal Cancer Liver Metastasis. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 2023, 27, 1498–1509. [Google Scholar] [CrossRef]
  22. Halazun, K.J.; Najjar, M.; Abdelmessih, R.M.; Samstein, B.; Griesemer, A.D.; Guarrera, J.V.; Kato, T.; Verna, E.C.; Emond, J.C.; Brown, R.S. Recurrence After Liver Transplantation for Hepatocellular Carcinoma. Ann. Surg. 2017, 265, 557–564. [Google Scholar] [CrossRef]
  23. Huang, A.; Guo, D.-Z.; Zhang, X.; Sun, Y.; Zhang, S.-Y.; Zhang, X.; Fu, X.-T.; Wang, Y.-P.; Yang, G.-H.; Sun, Q.-M.; et al. Serial Circulating Tumor DNA Profiling Predicts Tumor Recurrence after Liver Transplantation for Liver Cancer. Hepatol. Int. 2023, 18, 254–264. [Google Scholar] [CrossRef]
  24. Tie, J.; Cohen, J.D.; Lahouel, K.; Lo, S.N.; Wang, Y.; Kosmider, S.; Wong, R.; Shapiro, J.; Lee, M.; Harris, S.; et al. Circulating Tumor DNA Analysis Guiding Adjuvant Therapy in Stage II Colon Cancer. N. Engl. J. Med. 2022, 386, 2261–2272. [Google Scholar] [CrossRef]
  25. Powles, T.; Assaf, Z.J.; Davarpanah, N.; Banchereau, R.; Szabados, B.E.; Yuen, K.C.; Grivas, P.; Hussain, M.; Oudard, S.; Gschwend, J.E.; et al. ctDNA Guiding Adjuvant Immunotherapy in Urothelial Carcinoma. Nature 2021, 595, 432–437. [Google Scholar] [CrossRef]
  26. Liu, W.; Jin, K.-M.; Zhang, M.-H.; Bao, Q.; Liu, M.; Xu, D.; Wang, K.; Xing, B.-C. Recurrence Prediction by Circulating Tumor DNA in the Patient with Colorectal Liver Metastases After Hepatectomy: A Prospective Biomarker Study. Ann. Surg. Oncol. 2023, 30, 4916–4926. [Google Scholar] [CrossRef]
  27. Nishioka, Y.; Chun, Y.S.; Overman, M.J.; Cao, H.S.T.; Tzeng, C.-W.D.; Mason, M.C.; Kopetz, S.W.; Bauer, T.W.; Vauthey, J.-N.; Newhook, T.E.; et al. Effect of Co-Mutation of RAS and TP53 on Postoperative ctDNA Detection and Early Recurrence after Hepatectomy for Colorectal Liver Metastases. J. Am. Coll. Surg. 2022, 234, 474. [Google Scholar] [CrossRef]
  28. Kotani, D.; Oki, E.; Nakamura, Y.; Yukami, H.; Mishima, S.; Bando, H.; Shirasu, H.; Yamazaki, K.; Watanabe, J.; Kotaka, M.; et al. Molecular Residual Disease and Efficacy of Adjuvant Chemotherapy in Patients with Colorectal Cancer. Nat. Med. 2023, 29, 127–134. [Google Scholar] [CrossRef]
  29. Yoo, C.; Laliotis, G.; Jeong, H.; Jeong, J.H.; Kim, K.-P.; Lee, S.; Ryoo, B.-Y.; Sharma, S.; Dutta, P.; Malhotra, M.; et al. Utility of Circulating Tumor DNA (ctDNA) as a Predictive Biomarker for Disease Monitoring in Patients (Pts) with Cholangiocarcinoma (CCA) before and during Adjuvant Chemotherapy (ACT): Sub-Analysis of the Randomized Phase 2 STAMP Trial. J. Clin. Oncol. 2023, 41 (Suppl. S16), 4123. [Google Scholar] [CrossRef]
  30. Wang, J.; Huang, A.; Wang, Y.-P.; Yin, Y.; Fu, P.-Y.; Zhang, X.; Zhou, J. Circulating Tumor DNA Correlates with Microvascular Invasion and Predicts Tumor Recurrence of Hepatocellular Carcinoma. Ann. Transl. Med. 2020, 8, 237. [Google Scholar] [CrossRef] [PubMed]
  31. Jiang, N.; Zeng, X.; Tang, J.; Liang, Z.; Qi, X.; Huang, L.; Pang, F. Circulating Tumor DNA Is a Potential Prognostic Risk Factor of Recurrence in Patients with Hepatocellular Carcinoma Treated by Liver Transplantation. J. Clin. Oncol. 2022, 40 (Suppl. S16), e16196. [Google Scholar] [CrossRef]
  32. Chan, H.T.; Nagayama, S.; Otaki, M.; Chin, Y.M.; Fukunaga, Y.; Ueno, M.; Nakamura, Y.; Low, S.-K. Tumor-Informed or Tumor-Agnostic Circulating Tumor DNA as a Biomarker for Risk of Recurrence in Resected Colorectal Cancer Patients. Front. Oncol. 2023, 12, 1055968. [Google Scholar] [CrossRef] [PubMed]
  33. Ambrozkiewicz, F.; Trailin, A.; Červenková, L.; Vaclavikova, R.; Hanicinec, V.; Allah, M.A.O.; Palek, R.; Třeška, V.; Daum, O.; Tonar, Z.; et al. CTNNB1 Mutations, TERT Polymorphism and CD8+ Cell Densities in Resected Hepatocellular Carcinoma Are Associated with Longer Time to Recurrence. BMC Cancer 2022, 22, 884. [Google Scholar] [CrossRef]
  34. Tavolari, S.; Brandi, G. Mutational Landscape of Cholangiocarcinoma According to Different Etiologies: A Review. Cells 2023, 12, 1216. [Google Scholar] [CrossRef]
  35. Zhang, L.; Shay, J.W. Multiple Roles of APC and Its Therapeutic Implications in Colorectal Cancer. JNCI J. Natl. Cancer Inst. 2017, 109, djw332. [Google Scholar] [CrossRef]
  36. Lamlum, H.; Ilyas, M.; Rowan, A.; Clark, S.; Johnson, V.; Bell, J.; Frayling, I.; Efstathiou, J.; Pack, K.; Payne, S.; et al. The Type of Somatic Mutation at APC in Familial Adenomatous Polyposis Is Determined by the Site of the Germline Mutation: A New Facet to Knudson’s “two-Hit” Hypothesis. Nat. Med. 1999, 5, 1071–1075. [Google Scholar] [CrossRef]
  37. Wang, R.; Li, J.; Zhou, X.; Mao, Y.; Wang, W.; Gao, S.; Wang, W.; Gao, Y.; Chen, K.; Yu, S.; et al. Single-Cell Genomic and Transcriptomic Landscapes of Primary and Metastatic Colorectal Cancer Tumors. Genome Med. 2022, 14, 93. [Google Scholar] [CrossRef]
  38. Michel, M.; Kaps, L.; Maderer, A.; Galle, P.R.; Moehler, M. The Role of P53 Dysfunction in Colorectal Cancer and Its Implication for Therapy. Cancers 2021, 13, 2296. [Google Scholar] [CrossRef]
Figure 1. Timeline for ctDNA testing, cancer work-up, and surveillance as per institutional protocol. Note the example tumor marker shown is AFP for HCC; for other cancer types, the corresponding serum tumor marker (CCA: CA19-9, CRLM: CA19-9) is used for assessment.
Figure 1. Timeline for ctDNA testing, cancer work-up, and surveillance as per institutional protocol. Note the example tumor marker shown is AFP for HCC; for other cancer types, the corresponding serum tumor marker (CCA: CA19-9, CRLM: CA19-9) is used for assessment.
Cancers 16 00927 g001
Figure 2. Oncoprints of genomic alterations detected using Guardant 360 available for CCA post-transplant (A), HCC pre-transplant (B), HCC post-transplant (C), CRLM pre-transplant (D), and CRLM post-transplant (E). Type of genomic alteration represented by color with key at bottom of figure.
Figure 2. Oncoprints of genomic alterations detected using Guardant 360 available for CCA post-transplant (A), HCC pre-transplant (B), HCC post-transplant (C), CRLM pre-transplant (D), and CRLM post-transplant (E). Type of genomic alteration represented by color with key at bottom of figure.
Cancers 16 00927 g002
Table 1. Summary of demographic and pre-transplant variables and post-transplant outcomes.
Table 1. Summary of demographic and pre-transplant variables and post-transplant outcomes.
ALL
N = 21
HCC
N = 9
HCC/CCA
N = 1
CCA
N = 3
CRLM
N = 8
Male Sex, N (%)16 (76%)8 (89%)02 (67%)6 (75%)
Race, N (%)
White
Black
Other/Unknown

18 (86%)
2 (10%)
1 (5%)

8 (89%)
0
1 (11%)

1 (100%)
0
0

2 (50%)
1 (25%)
0

7 (88%)
1 (13%)
0
Age at Transplant Surgery, Median (IQR)55 (50–68)70 (46–73)6051 (25–55)54 (49–60)
Cirrhosis, N (%)
Non-Malignancy Cirrhosis Factors
A1AT
ETOH
HBV
HCV
NASH
PSC
Biliary Atresia
Chemotherapy-Induced
PBC
19 (90%)

1 (5%)
2 (10%)
3 (14%)
1 (5%)
6 (29%)
2 (10%)
2 (10%)
1 (5%)
1 (5%)
9 (100%)

0
1 (11%)
2 (22%)
1 (11%)
5 (56%)
0
2 (22%)
0
0
1 (100%)

1 (100%)
0
1 (100%)
0
0
0
0
0
0
3 (75%)

0
1 (25%)
0
0
0
1 (25%)
0
0
0
6 (86%)

0
0
0
0
1 (14%)
1 (14%)
0
1 (14%)
1 (14%)
MELD Score, Median (IQR)15 (11–24)22 (14–25)2412 (10–29)11 (7–19)
Pre-Treatment Tumor Marker Level, Mean (SD)
AFP (ng/mL)
CA19-9 (U/mL)
CEA (ng/mL)

8 (6, 14)
23 (12, 168)
31 (1, 64)

8 (6, 13)


39
216


22 (8, 24)



31 (1, 64)
Pre-Transplant Number of Lesions, N (%)
1
2–3
Innumerable

11 (52%)
6 (29%)
2 (10%)

7 (78%)
1 (11%)
0

0
1 (100%)
0

4 (100%)
0
0

0
4 (50%)
2 (25%)
Pre-Transplant Size of Biggest Lesion (cm), Median (IQR)4 (2–6)1 (1–4)316.7 (3–8)
Pre-Transplant Treatment, N (%)
Systemic Chemotherapy
Radiotherapy
SBRT
Prior Surgery
Ablation
Chemoembolization
Radioembolization
Immunotherapy
18 (86%)
10 (48%)
6 (29%%)
4 (19%)
3 (14%)
4 (19%)
6 (29%)
5 (24%)
1 (5%)
7 (78%)
0
0
0
0
1 (11%)
2 (22%)
4 (44%)
0
0
0
0
0
0
0
0
0
0
3 (100%)
2 (67%)
2 (67%)
2 (67%)
0
0
0
0
0
8 (100%)
8 (100%)
4 (50%)
2 (25%)
3 (38%)
3 (38%)
4 (50%)
1 (13%)
1 (13%)
Post-Transplant Tumor Marker Level, Median (IQR)
AFP (ng/mL)
CA19-9 (U/mL)
CEA (ng/mL)

3 (3–7)
13 (6–20)
2 (1–2)

3 (3–6.8)


4.8



13 (6–20)



1.7 (1–2)
Recurrence, N (%)6 (29%)3 (33%)01 (25%)2 (25%)
Patient Status, N (%)
Alive
Dead

17 (81%)
4 (19%)

6 (67%)
3 (33%)

1 (100%)
0

2 (67%)
1 (33%)

8 (100%)
0
Cancer-Related Deaths, N (%)2 (10%)2 (22%)000
Recurrence Survival (Days), Median (IQR)
Overall Survival (Days), Median (IQR)
13 (7–28)
14 (8–39)
12 (5–31)
14 (6–34)
16.7
16.7
8 (6–15)
8 (6–32)
14 (10–40)
25 (10–60)
Key: A1AT = alpha-1-anti-trypsin deficiency, ETOH = ethanol, HBV = hepatitis B virus, HCV = hepatitis C virus, NASH = non-alcoholic steatohepatitis, PSC = primary sclerosing cholangitis, PBC = primary biliary cholangitis, AFP = alpha-fetoprotein, CA19-9 = cancer antigen 19-9, CEA = carcinoembryonic antigen, SBRT = stereotactic body radiation therapy.
Table 2. Transplant variables.
Table 2. Transplant variables.
PtAgeSexCancer TypeCirrhosis FactorsMELD at TxTx TypeLiver Transplant TechniqueAberrant Liver VasculatureType of Arterial AnastomosisType of Venous AnastomosisBiliary AnastomosisReal Warm Ischemia Time (min)LT Duration (min)RBCs, FFP (Units)Reperfusion OrderPost-Reperfusion Syndrome
139MHCCBiliary atresia, PBC12LDLTPiggyback-StandardInterpositionHJ387760, 0Vein firstNo
270MHCCNASH22DCDPiggyback-Standard End-to-endDuct-to-duct435416, 3Vein firstNo
352MHCCHCV23DCDConventionalReplaced RHAStandardEnd-to-endDuct-to-duct---Vein firstYes
475MHCCNASH25DBDPiggyback-StandardEnd-to-endDuct-to-duct465052, 0Vein firstNo
566MHCCNASH, ETOH, HBV9LDLTPiggybackAccessory LHAStandard End-to-endDuct-to-duct397200, 0Vein firstNo
670MHCCHBV25DBDPiggybackAccessory RHAStandard End-to-endDuct-to-duct464300, 0Vein firstNo
772MHCCNASH11LDLTPiggyback-Standard End-to-endDuct-to-duct425577, 5Vein firstNo
873MHCCNASH12DCDPiggyback-Standard End-to-endDuct-to-duct584104, 1BothNo
932FHCCBiliary atresia40Split, DBDConventional-Standard End-to-endHJ4565718, 13Vein firstNo
1060FHCC/CCAHBV, A1AT24DCDPiggyback-Standard End-to-endDuct-to-duct483858, 1BothYes
1125MCCAPSC12DCDConventional-Standard End-to-endHJ424903, 0Vein firstYes
1251FCCAETOH29DBDPiggybackReplaced LHAStandard ConduitHJ2768317, 11Vein firstYes
1355MCCA-10DBDConventionalReplaced RHAInfra-renalConduitHJ404521, 0Vein firstNo
1450MCRLM-6LDLTPiggyback-StandardEnd-to-endDuct-to-duct395840, 0Vein firstNo
1553MCRLM-6DBDConventional-Standard End-to-endHJ494270, 0Vein firstYes
1661MCRLM-13LDLTConventional-Standard End-to-endDuct-to-duct328694, 0Vein firstNo
1764MCRLM-11LDLTPiggyback-Standard End-to-endDuct-to-duct435947, 8Vein firstNo
1854MCRLM-14LDLTPiggyback-Standard InterpositionDuct-to-duct679925, 0Vein firstNo
1949FCRLMPBC23LDLTPiggyback-Standard End-to-endDuct-to-duct276852, 0Vein firstNo
2049MCRLM-21DBDPiggyback-Standard End-to-endHJ3970820, 12Vein firstNo
2156FCRLMNASH8LDLTPiggyback-Standard InterpositionDuct-to-duct457004, 0Vein firstYes
Key: PBC = primary biliary cholangitis, NASH = non-alcoholic steatohepatitis, HCV = hepatitis C virus, ETOH = ethanol, HBV = hepatitis B virus, A1AT = alpha-1 anti-trypsin deficiency, PSC = primary sclerosing cholangitis, LDLT = living donor liver transplant, DCD = donation after cardiac death, DBD = donation after brain death, RHA = right hepatic artery, LHA = left hepatic artery, HJ = hepaticojejunostomy.
Table 3. Post-transplant variables.
Table 3. Post-transplant variables.
PatientInduction ISInitial ISIS 12 monthBiliary ComplicationsBiliary InterventionArterial Complications, InterventionAcute Rejection GradeTreatment of Acute RejectionChronic Rejection
1BasiliximabGC + Tacrolimus-LeakPTHC----
2-GC + MMF + Tacrolimus-------
3BasiliximabGC + TacrolimusTacrolimus + Sirolimus------
4BasiliximabGC + MMF + TacrolimusCyclosporine + Everolimus------
5BasiliximabGC + MMF + TacrolimusTacrolimus + MMFLeakERCP-MildIV steroids-
6BasiliximabGC + MMF + TacrolimusTacrolimus + MMF + Sirolimus------
7BasiliximabGC + MMF + Tacrolimus-------
8-GC + MMF + TacrolimusTacrolimus + EverolimusStrictureERCP----
9-GC + MMF + Tacrolimus-------
10BasiliximabGC + MMF + TacrolimusTacrolimus + Everolimus------
11-GC + MMF + Tacrolimus-Leak, ischemic cholangiopathy HJ reconstruction, PTHC, re-transplantHA stenosis and pseudoaneurysm, stent placement---
12BasiliximabGC + MMF + TacrolimusTacrolimus + GC + MMF------
13-GC + MMF + Tacrolimus-------
14BasiliximabGC + MMF + Tacrolimus-------
15-GC + MMF + TacrolimusTacrolimus + EverolimusLeakRe-operation----
16BasiliximabGC + MMF + Tacrolimus-StrictureERCP-MildIV steroids-
17BasiliximabGC + MMF + TacrolimusTacrolimus + EverolimusLeakERCP + PTC----
18BasiliximabGC + MMF + TacrolimusTacrolimus + EverolimusStrictureERCP----
19-GC + MMF + Tacrolimus-------
20-GC + MMF + Tacrolimus-------
21-GC + MMF + Tacrolimus-------
Table 4. Tumor marker correlation with ctDNA testing at times prior to and following transplant, along with time of recurrence.
Table 4. Tumor marker correlation with ctDNA testing at times prior to and following transplant, along with time of recurrence.
PtCancer TypeDate Liver Cancer dxDx Date Tumor Marker LevelDx Tumor Marker LevelDate Pre-Transplant MarkerPre-Transplant Tumor MarkerDate of Pre-Transplant ctDNAPre- ctDNA ResultsDate of TransplantDate Post-Transplant MarkerPost-Transplant Tumor MarkerDate of Post-Transplant ctDNAPost-Transplant ctDNA ResultsctDNA TimingDate of RecurrenceDate of Tumor Marker Level with RecurrenceRecurrence Tumor Marker Level
1HCC7/20228/2/2023AFP: 158/15/23AFP: 24.18/15/23+ (CDx)8/21/2310/26/23AFP: <3.0
2HCC2/16/20234/17/2023AFP: <34/17/2023AFP: <34/19/2023+ (CDx)6/9/202312/5/23AFP: <3.06/20/23+Both
3HCC8/10/20108/18/2010AFP: 8.79/24/2010AFP: 4.6 10/12/201010/21/2010AFP:7.112/19/2019+Post-12/16/201912/17/2019AFP: 4398.6
4HCC12/18/202012/18/2020AFP: 6.23/7/2022AFP: 9.3 4/9/20226/21/2022AFP: <3.0 4/11/2023+Post-
5HCC7/11/20221/27/2022AFP: 111/27/2022AFP: 11 7/11/20227/28/2022AFP: <3.0 8/10/2022+Post-
6HCC2/18/20192/18/2019AFP: 7.62/3/2020AFP: 4.9 5/1/202010/23/2020AFP: <311/12/2021+Post-10/23/20206/2/21AFP: <3.0
7HCC3/15/20223/15/2022AFP: 7.1 09/08/2022 AFP: 9.5 9/18/202210/10/2022AFP: 10.511/14/2022+Post-10/4/202310/4/23AFP: <3.0
8HCC11/8/20198/2/2019AFP: 5.62/20/2020AFP: 6.312/18/2019- 4/12/202012/16/2020AFP: <3 Pre-
9HCC7/19/20227/19/2022AFP: 1412/8/2022AFP: 8.18/15/2022+12/30/202211/18/2022AFP: 69/1/2023-Both
10HCC/CCA5/4/20215/4/2021; 5/20/2021AFP: 38.8, CA19-9: 2167/5/2022AFP: 36.6, CA 19-9: 8346/6/2022+7/7/20227/22/2022AFP: 4.8 Pre-
11CCA7/14/20216/9/2021CA19-9: 81/12/2023CA 19-9: 467/20/2022, 9/2/22+, +2/7/2023, 07/13/23 9/28/2023-Both
12CCA7/3/20206/19/2020CA19-9: 221/10/2023CA 19-9: 15 3/1/2023 7/31/2023 CA19-9: 6.36/14/2022+Post-11/3/202110/25/21; 5/25/21CA 19-9: 146; AFP: <3
13CCA11/25/20221/21/2021CA19-9: 248/4/2020CA 19-9: 4512/13/2022-8/6/202012/1/2020CA19-9: 208/1/2023-Both
14CRLM6/201712/10/2018CEA: 2.49/21/22CEA: 110/27/22, 9/25/23-, -10/11/2312/11/23CEA: 1.611/1/23+Both
15CRLM2/20/20203/3/2020CEA: 68548/9/2022CEA: 4.95/19/2022+9/14/20221/10/2023CEA: 1.811/15/2022- (GR)Both
16CRLM10/5/20179/15/2017CEA: 60.11/6/2020CEA: 10.411/11/2019+1/12/20202/6/2020CEA: 1.21/12/2022, 7/15/22, 1/16/23-, -, - (GR)Both
17CRLM20198/26/2021CEA: 110/28/2022CEA: 2.711/1/2022+11/1/202212/15/2022CEA: 0.912/8/2022, 6/7/23+, +Both
18CRLM11/12/20118/23/2011CEA: 1.69/10/2020CEA: 1.6 6/25/2019+9/13/20201/9/2023CEA: 1.711/8/2021, 5/5/22- (GR)Both
19CRLM4/1/20164/14/2016CEA: 64.411/27/2017CEA: 3.7 4/22/20185/24/2018CEA: 3.81/23/2020, 4/19/22+, +Post-
20CRLM6/16/2012N/AN/A5/27/2018CEA: 2.9 5/27/20188/30/2018CEA: 25/27/2022, 2/23/22, 7/28/23-, -, - (GR)Post-9/19/20199/19/2019CEA: 1.8
21CRLM11/9/202011/2/2020CEA: 30.78/9/2022 8/6/2022, 11/10/22+, -2/6/20237/3/23CEA: 16.510/31/23+Both9/25/237/17/23; 9/25/23CEA: 17.2; CEA: 17.9
Key: CDx = Guardant CDx; GR = Guardant Reveal. All other ctDNA results are from Guardant360. AFP units = ng/mL, CA19-9 units = U/mL, CEA units = ng/mL. “+” and “-” symbols correspond to presence or absence of ctDNA respectively.
Table 5. Oncologic variables including treatment before and after liver transplant as well as with recurrence.
Table 5. Oncologic variables including treatment before and after liver transplant as well as with recurrence.
PtCancer TypeLiver Cancer dxPre-Transplant TreatmentChemotherapy DetailsRadiation Therapy DetailsSurgery DetailsPathologic ResponseDate of TransplantDate of RecurrenceRecurrence, Number of Tumors, SitesLargest Tumor Size (cm)Treatment of RecurrenceRecurrence Treatment DetailsDeath, Cause
1HCC07/2022TARE 9/2022 PR8/15/23
2HCC2/16/2023- 6/9/2023 No
3HCC8/10/2010Microwave ablation 10/12/201012/16/2019Intrahepatic; multifocal11.5Chemotherapy02/2/2020–11/6/2020: levatinib; switched to cobozanrinib after progression until 11/6/202012/17/2020; HCC
4HCC12/18/2020TACE, TARE03/2/12, 5/11/21, 8/18/211/14/22 PR4/9/2022 No
5HCC7/11/2022- 7/11/2022 No
6HCC2/18/2019TARE 09/19/2019, 11/19/2019 SD5/1/202010/23/2020Extrahepatic; multifocal—lung, adrenal fossa, retrocaval lymph nodes1.3Chemotherapy, radiation9/7/21: radiation; 12/3/21–2/3/22: levatinib5/13/2022: HCC
7HCC3/15/2022TARE 5/18/22 SD9/18/202210/4/2023Extrahepatic; multifocal—lung1.5Chemotherapy11/22/23: levatinib
8HCC11/8/2019TACE12/2/2019 PR4/12/2020 No 10/8/2023: metastatic melanoma
9HCC7/19/2022SBRT09/26/22–10/10/22: 4 treatments PR12/30/2022 No
10HCC/CCA5/4/2021- 7/7/2022 No
11CCA7/14/2021Chemoradiation08/29/22–09/16/22: capecitabine08/29/22–09/16/22 SD2/7/2023, 07/13/23 No
12CCA7/3/2020SBRT09/26/2019–09/27/2019 CR8/6/202011/3/2021Extrahepatic; multifocal—liver, bone Chemotherapy, radiation9/6/22–9/21/22: radiation; 12/1/21–7/1/22: gemcitabine/oxaliplatin; 7/26/22–8/1/22: FOLFIRI; 10/1/22–12/1/22: gemcitabine/abraxane x 3 with PR3/15/23: cardiovascular event during dialysis; CCA
13CCA11/25/2022Chemoradiation, SBRT1/10/23–2/3/23: capecitabine1/10/23–2/3/23 CR3/1/2023, adjuvant capecitabine x 4 cycles (6/5/23) No
14CRLM2015Chemotherapy, surgery, microwave ablation7/11/17–9/20/17, 8/2019–5/8/2018: FOLFOX/cetuximab; 8/19–2/20: capecitabine, 8 cycles; 4/19/21–9/21: capecitabine5/18/22: microwave ablation; 2/2/23: SBRT 30 Gy in 1 fraction12/12/2017: open wedge resection (segments 4–8); 1/18/19: segment 4b lesion resection; 7/2/19: segment 8 lesion resection; 2/23/21: segments 7/8 liver resectionCR10/11/23 No
15CRLM2/20/2020Chemotherapy, immunotherapy, radiation therapy3/20–8/18/20: CAPOX, bevacizumab; 10/2020–early 2021: 5FU, bevacizumab; 07–08/21: 5FU only; 10/21–01/22: 5FU, bevacizumab01–06/2021 CR9/14/2022 No
16CRLM10/5/2017Chemotherapy, TARE10/2017–02/2018: FOLFOX, Avastin x 9 cycles; 02/18–12/11/19: FOLFIRI/panitumumab4 rounds PR1/12/2020 No
17CRLM2019Chemotherapy, radiation therapy, SBRT09–11/11/2020: FOLFOX, Avastin x 12 cycles; 12/2020–05/2021: AvastinSBRT: 9/20/2020 CR11/1/2022 No
18CRLM11/12/2011Chemotherapy, radiation therapy, surgery, TACE, RFA10/18/2011–04/2012: Xeloda, FOLFIRI x 3 cycles; 07/2017: FOLFIRI, Erbitux; 02/25/15–03/2015: HAI pump infusion therapy Hepatic resection 02/25/2015 and 09/2016PR9/13/2020 No
19CRLM4/1/2016Surgery,
TACE, chemotherapy
1/17/2015: HAI FUDR; 8/26/2016: FOLFIRI w/ panitumumab x 6 cycles, FOLFOX Avastin x 3 cycles Wedge resection segments 2 and 3, caudate lobe removal, R hepatectomyCR4/22/2018 No
20CRLM6/16/2012Chemotherapy, ablation, TACE, radiotherapy08–10/2013: FOLFIRI; 12/2013: hepatic resection, HAI pump; until 10/2014: FUDR; 01–04/2014: 5FU; 05–01/2016: irinotecan, cetuximab; 02/2016–11/2017: 5FU cetuximab, 3/7/2018: FOLFOX x 13 cycles12/2017: proton beam radiotherapy07/2013: Ablation*5/27/20189/19/2019Extrahepatic; unifocal, right upper lobe of lung0.9Chemotherapy, surgeryRight upper lobe metastectomy; 12/16/2019–7/27/2020: FOLFIRI, bevacizumab with complete response
21CRLM11/9/2020Chemotherapy, TACE5/2021: FOLFOX x 7 cycles; 6/28/22–11/7/22: irinotecan; 9/28/22–1/4/23: panitumumab; 3/2/22: infusional 5FU PR2/6/239/25/23Intrahepatic and extrahepatic—lung nodule Chemotherapy, plan for surgery10/17/23: irinotecan, panitumumab
Key: SBRT = stereotactic radiation body therapy, TACE = trans-arterial chemoembolization, TARE = trans-arterial radioembolization, RFA = radiofrequency ablation, PR = partial response, CR = complete response, SD = stable disease. * = Information at OSH.
Table 6. Pre- vs. post-transplant mutational profiles of patients who underwent sequential ctDNA testing by cancer type.
Table 6. Pre- vs. post-transplant mutational profiles of patients who underwent sequential ctDNA testing by cancer type.
Patient #Cancer TypeTime From Pre-op Testing to Surgery (Days)Pre-op Somatic Alterations DetectedPre-Transplant ctDNATime from Surgery to Post-op Testing (Days)Post-op Somatic Alterations DetectedPost-Transplant ctDNA
2HCC51YesCTNNB1 L31V 0.20%11YesCTNNB1 D32V N/A
9HCC137YesTERT Promoter SNV 0.80%
FGFR2 K509E 2.00%
245NoNot Identified
11HCC148YesNot Identified233NoNot Identified
13CCA78NoNot Identified153NoNot Identified
14CRLM16NoNot Identified21YesROS1 L1899F 0.2%
15CRLM26YesMTOR Q1715 0.40%62NoNot Identified
16CRLM62YesAPC E1064 * 0.50%
TP53 R248Q 0.10%
SMAD4 A418fs 0.06%
MAP2K1 K84R 0.20%
731NoNot Identified
17CRLM0YesNF1 A706V 0.10%
MLH1 I191 0.20%
37YesFGFR3 T317A 1.80%
PALB2 N241D 1.60%
BRCA2 C1290Y 1.50%
ROS1 T632N 1.20%
MET V378I 0.10%
18CRLM293YesROS1 A2106T 0.20%
BRCA1 K22E 0.10%
421NoNot Identified
21CRLM184YesAPC S1415fs 1%
TP53 S149fs 1.3%
266YesAPC S1415fs 0.2%
TP53 S149fs 0.2%
Note: Percentages shown represent %cfDNA (cell-free DNA). N/A = not available. Asterisk (*) indicates unknown substitution.
Table 7. Tumor details from diagnostic radiologic imaging and explant pathology.
Table 7. Tumor details from diagnostic radiologic imaging and explant pathology.
PtCancer TypeDxNumber of TumorsDx-Largest Tumor Size (cm)Pathologic Tumor NumbersPathologic Largest Tumor Size (Viable) (cm)% Viable Tumor Explanted LiverPathologic Vascular InvasionPathologic Perineural InvasionPathologic Liver Capsule InvolvementHistologic Grade of DifferentiationMSIPathologic TNM Staging from Transplant
1HCC13.630.820%Small vesselAbsentAbsentG2 T2
2HCC16.612.5100%AbsentAbsentAbsentG2 T1b
3HCC14130%AbsentAbsentAbsentG2 T1bN0
4HCC32.812.3100%AbsentAbsentAbsentG2 T2
5HCC51.751.7100%Small vesselAbsentAbsentG2–3 T2
6HCC14.942.75%Small vesselAbsentAbsentG2 T2N0
7HCC14.2Multiple4.350%Small and large vesselAbsentAbutsG2 T4
8HCC12.610.850%AbsentAbsentAbsentG2 T1a
9HCC1812.320%AbsentAbsentAbsentG2 T1b
10HCC/CCA32.33 (2-HCC, 1-CCA)2-HCC, 10-CCA0%, 5%, 95%PresentPresentPosterior capsuleG2–3 T2
11CCA1110.1100%AbsentAbsentAbsentG1 T2aN0
12CCA1100N/AN/AN/AN/AN/A
13CCA111 (residual)No gross lesion visible G2 T1N0
14CRLMNumerous7.600N/AN/AN/AN/AN/AStableT0N1aM1
15CRLMNumerous7.7214.120%AbsentAbsentAbsent StableT3N1M1a
16CRLM35.834100%, 0%AbsentAbsentAbsent StableT3N1aM1
17CRLM**18.50%AbsentAbsentAbsent StableT3N1aM1
18CRLM3*140%AbsentAbsentAbsent Stable
19CRLM2*0 StableT3N1aM1
20CRLM**41.7100%AbsentAbsentAbsentG2Unknown
21CRLM21.463.3100%AbsentAbsentAbsentG2StableT3N0M1
Key: * = imaging performed at OSH, Dx = diagnostic, N/A = not applicable.
Table 8. ctDNA profiles for patients who experienced recurrence.
Table 8. ctDNA profiles for patients who experienced recurrence.
Patient NumberCancer TypeDate
Pre-Transplant ctDNA Collected
Pre-op Somatic Alterations
Detected
Pre-Transplant ctDNADate
Post-Transplant ctDNA Collected
Post-op Somatic
Alterations Detected
Post-Transplant ctDNADate of Recurrence
3HCC 12/19/2019YesCTNNB1 T41A 3.70%
TERT Promoter 2.00%
12/16/2019
6HCC 11/12/21YesARID1A S696fs 0.70%
CTNNB1 S33A 16.50%
TERT promoter 13.30%
10/23/2020
7HCC 11/14/2022YesTP53 R248Q 0.10%
FGFR1 V247V 6.00%
10/4/2023
12CCA12/3/22NoNot identified8/1/23NoNot identified11/3/2021
20CRLM 7/28/23NoNot Identified9/19/2019
21CRLM8/6/22YesTP53 S149fs 1.30%
APC S1415fs 1.00%
AR R780W 0.50%
10/31/23YesAPC S1415fs 0.2%
TP53 S149fs 0.2%
9/25/23
Note: Percentages shown represent %cfDNA (cell-free DNA).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hong, H.; Wehrle, C.J.; Zhang, M.; Fares, S.; Stitzel, H.; Garib, D.; Estfan, B.; Kamath, S.; Krishnamurthi, S.; Ma, W.W.; et al. Circulating Tumor DNA Profiling in Liver Transplant for Hepatocellular Carcinoma, Cholangiocarcinoma, and Colorectal Liver Metastases: A Programmatic Proof of Concept. Cancers 2024, 16, 927. https://doi.org/10.3390/cancers16050927

AMA Style

Hong H, Wehrle CJ, Zhang M, Fares S, Stitzel H, Garib D, Estfan B, Kamath S, Krishnamurthi S, Ma WW, et al. Circulating Tumor DNA Profiling in Liver Transplant for Hepatocellular Carcinoma, Cholangiocarcinoma, and Colorectal Liver Metastases: A Programmatic Proof of Concept. Cancers. 2024; 16(5):927. https://doi.org/10.3390/cancers16050927

Chicago/Turabian Style

Hong, Hanna, Chase J. Wehrle, Mingyi Zhang, Sami Fares, Henry Stitzel, David Garib, Bassam Estfan, Suneel Kamath, Smitha Krishnamurthi, Wen Wee Ma, and et al. 2024. "Circulating Tumor DNA Profiling in Liver Transplant for Hepatocellular Carcinoma, Cholangiocarcinoma, and Colorectal Liver Metastases: A Programmatic Proof of Concept" Cancers 16, no. 5: 927. https://doi.org/10.3390/cancers16050927

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop