Next Article in Journal
Differences in Internet Use and eHealth Needs of Adolescent and Young Adult Versus Older Cancer Patients; Results from the PROFILES Registry
Previous Article in Journal
CRISPR-to-Kill (C2K)–Employing the Bacterial Immune System to Kill Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liver Transplantation for Hepatocellular Carcinoma after Downstaging or Bridging Therapy with Immune Checkpoint Inhibitors

Department of Surgery, Duke University Medical Center, Durham, NC 27705, USA
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(24), 6307; https://doi.org/10.3390/cancers13246307
Submission received: 23 October 2021 / Revised: 13 December 2021 / Accepted: 14 December 2021 / Published: 15 December 2021

Abstract

:

Simple Summary

Immune checkpoint inhibitors (ICI) have revolutionized the treatment of hepatocellular carcinoma (HCC). In addition to their role in advanced HCC, there is considerable interest in using ICIs in the neoadjuvant setting, either as a downstaging or bridging therapy, prior to potentially curative liver transplantation. In this article, we reviewed all the available literature on ICI use in this context. We postulate that ICIs may be utilized safely prior to liver transplant; however, further research is needed in this area.

Abstract

Liver transplantation offers excellent outcomes for patients with HCC. For those who initially present within the Milan criteria, bridging therapy is essential to control disease while awaiting liver transplant. For those who present beyond the Milan criteria, a liver transplant may still be considered following successful downstaging. Since the introduction of atezolizumab as part of the first-line treatment for HCC in 2020, there has been increasing interest in the use of ICIs as bridging or downstaging therapies prior to liver transplant. A total of six case reports/series have been published on this topic, with mixed outcomes. Overall, liver transplantation can be performed safely following prolonged ICI use, though ICIs may increase the risk of fulminant acute rejection early in the post-operative period. A minimal washout period between the last dose of ICI and liver transplantation has been identified as an important factor predicting transplant outcomes; however, further research is needed.

1. Introduction

Liver cancer is the sixth most common cancer worldwide, with hepatocellular carcinoma (HCC) accounting for 90% of cases [1]. Surgical resection and liver transplantation have been the cornerstones of early-stage HCC treatment. The decision to resect or transplant is influenced by many factors, such as tumor characteristics, the presence or absence of portal hypertension, the severity of underlying liver dysfunction, and patient performance status, thus remaining a topic of debate [2]. In general, hepatic resection is preferred among HCC patients with solitary lesions in whom perioperative hepatic decompensation is unlikely, whereas liver transplantation is the treatment of choice among those with multi-focal disease and more advanced liver dysfunction.
The Milan criteria are the primary selection criteria when evaluating the HCC tumor burden in patients being considered for liver transplant [3]. For patients with tumors within the Milan criteria, liver transplantation offers excellent short-term and long-term outcomes, with a 5-year overall survival over 70% and recurrence rate around 10–15% [4,5]. With careful patient selection, similar short-term outcomes can be achieved with tumor resection; however, the risk of recurrence at five years approaches 70% [6]. Furthermore, 10-year survival after liver resection for HCC is as low as 15% [7].
For patients with tumors beyond the Milan criteria, or beyond the intermediate stage based on the Barcelona Clinic Liver Cancer (BCLC) system, locoregional and systemic therapy have become the mainstay treatment options. The last decade has witnessed significant advancements in HCC systemic therapy. Sorafenib, approved in 2008, had previously been the only first-line treatment for advanced HCC [8]. In 2020, atezolizumab, a PD-L1 monoclonal antibody, plus bevacizumab, an anti-VEGF monoclonal antibody, have been shown to improve progression-free survival compared to sorafenib [9]. More importantly, this combination induced a complete response in 5.5% patients compared to 0% in the sorafenib arm of the study. The improvement in systemic therapy for HCC opens the unique possibility of treating HCC in a neoadjuvant context before liver transplantation.
In this review, we present the available literature on the clinical experience with ICIs in the neoadjuvant setting, either as a bridging or downstaging therapy for HCC before liver transplantation. Although there is a paucity of literature in this area, checkpoint inhibitors have been utilized safely prior to transplant. This review highlights potential directions for future research regarding the use of ICIs in HCC treatment.

2. Current Bridging and Downstaging Strategies Prior to Liver Transplantation

HCC is unique in that 80–90% cases develop in patients with underlying liver disease. Conceptually, liver transplant represents the ideal therapy, as both the primary tumor is resected and the risk for developing future liver neoplasms is significantly reduced. Unfortunately, the ongoing scarcity of donor organs has limited the utilization of transplantation for HCC to candidates with early-stage disease in an effort to avoid futility [10]. However, since the inception of the Milan criteria, efforts to refine and safely expand selection criteria have been developed and implemented in the field of transplant oncology [11,12,13,14,15,16,17]. As the listing criteria for transplants become less restrictive, the risk of tumor recurrence inevitably increases. For example, the Extended Toronto Criteria offer liver transplantation for patients with any number of tumors of all sizes, barring any extrahepatic disease, vascular invasion, or poorly differentiated status on biopsy. Even though the overall 10-year survival was not statistically different compared to those within the Milan criteria, the recurrence rate after transplantation in this population was close to 30% [16].
On a practical level, the shortage of suitable donor livers remains a major limitation to the field of transplantation. In the US transplant system, in which patients must meet Milan criteria to qualify for MELD exception points, many patients spend months to years on the waitlist. In the US, the median waiting time for candidates with MELD scores 15–34 (the vast majority of HCC patients fall within this category) was 5.6 months in 2019. Moreover, all HCC patients must now demonstrate disease stability for 6 months prior to receiving initial exception points [18]. As such, the transplant rate for patients with HCC exception points is over 50% lower than it was 10 years ago [19]. Within this framework, waitlist mortality and dropout rate are substantial. Of all patients listed for liver transplant in 2016, 11.3% died on the waitlist and an additional 23.3% were removed from the list over a 3-year period. Left untreated, the risk of dropout may approach 25% at 1 year [20].
Bridging therapies, designed to control or delay disease progression, are key for patients on the waitlist. Locoregional therapy (LRT) is the most common form of bridging therapy. LRT is an umbrella term that encompasses a range of therapies, such as radiofrequency ablation (RFA), microwave ablation (MWA), transarterial chemoembolization (TACE), radioembolization (Y-90), and stereotactic body radiotherapy [21]. There have been no randomized trials conducted evaluating the efficacy of LRT as a bridging therapy, and most retrospective comparative studies are subject to bias. Those selected for LRT are more likely to have more advanced tumors. As a result, among those who undergo liver transplantation, LRT bridge therapy has not been shown to improve overall survival or reduce recurrence risk [22,23]. However, because the dropout rate from the transplant waitlist can be reduced by LRT bridge therapy [24], LRT is now accepted as the standard of care for those expected to remain on waitlist for more than 6 months [25].
In addition to treating patients on the waitlist with HCC within the Milan criteria, LRT can be utilized as a downstaging therapy for patients initially presenting with HCC beyond the Milan criteria. For patients who are otherwise not eligible for a potentially curative liver transplant, the ability to effectively downstage patients confers a definite advantage regarding access to transplantation. Many retrospective studies have showed that HCC can be downstaged to within the Milan Criteria with LRT, and if downstaging is successful, long-term recurrence free survival and overall survival following transplant are comparable to those within the Milan Criteria [4,26,27,28,29]. The 2018 American Association for the Study of Liver Diseases (AASLD) guidelines now recommend the consideration of patients beyond the Milan criteria for transplant after successful downstaging to within the Milan criteria [30]. In 2017, UNOS/OPTN adopted an expanded set of inclusion criteria to allow the prioritization of those who are successfully downstaged [4]. The first prospective randomized trial to evaluate the benefit of liver transplantation after successful downstaging with LRT to within the Milan criteria was published in 2020. Out of the 74 patients enrolled, 61% were successfully downstaged and randomized. The 5-year tumor-free survival and overall survival were 76.8% and 77.5% for those who received liver transplantation, compared to 18.3% and 31.2% respectively for those who received non-transplantation therapies [31].

3. Immune Checkpoint Inhibitors for HCC

Immune checkpoint inhibitors (ICIs) have revolutionized the field of oncology in recent years. Their success has been replicated for multiple types of cancers, and there are currently numerous ongoing clinical trials designed to evaluate this new class of cancer therapy in different settings. ICIs are particularly attractive for HCC treatment for several reasons: (1) HCC responds poorly to traditional cytotoxic chemotherapies; (2) HCC evolves in the context of underlying chronic liver inflammation, which can lead to exhaustion and the immune escape of tumor cells; (3) the liver harbors a large repertoire of resident immune cells; (4) the liver is considered relatively immuno-privileged and thus tumor immunity is suppressed to some extent. Although there are many checkpoint receptor targets, such as CTLA-4, PD-1, TIM-3, VISTA, and OX-40, most therapies in clinical trials or in current use target CTLA-4, PD-1, and PD-L1. Various agents have been tested in HCC patients in a range of settings.
For early-stage HCC (BCLC stage 0 and A), tumor resection and ablation are the standard of care. Although no ICI treatment combinations have been approved, many are being tested in phase I–III trials in the adjuvant setting [32]. For intermediate-stage HCC (BCLC stage B), the standard of care is TACE or Y90. Although studies that have combined sorafenib and TACE over the years have not shown any added benefits [33,34], there is considerable interest in exploring the potentially synergistic effect between LRT and ICI. Theoretically, transarterial therapies lead to tumor necrosis and the release of tumor antigens, which, in the presence of checkpoint inhibition, can boost antitumor immunity. Additionally, studies demonstrate that TACE may enhance PD-1 expression on intratumor inflammatory cells and PD-L1 expression on HCC tumor cells [35].
For patients with advanced HCC (BCLC stage C) or those with intermediate HCC (BCLC stage B) who either are not a candidate for LRT or who have progressed despite therapy, treatment options are limited. sorafenib was the only first-line therapy approved for HCC until 2018, when lenvatinib, another tyrosine kinase inhibitor, was approved following a phase III non inferiority trial [36]. Most clinical trials on ICIs in HCC have focused on this group of patients. The IMbrave150 trial compared the atezolizumab and bevacizumab combination in advanced HCC patients to standard-of-care sorafenib and found better progression free survival and overall survival among those who received the atezolizumab-bevacizumab combination [9]. This ultimately led to the approval of atezolizumab-bevacizumab as a first-line treatment for advanced HCC. Similarly, three additional ICI therapies—nivolumab monotherapy, pembrolizumab monotherapy and nivolumab/ipilimumab dual therapy—have been approved by the FDA as second-line therapies based on published phase I–II trial results [37,38,39]. Furthermore, a number of ongoing trials are evaluating other ICI agents either as monotherapies or in combination with other treatment strategies [40].
Thus far, the toxicity of ICI agents appears to be limited. ICI-induced hepatoxicity (ICH) is a distinct entity and often presents as asymptomatic elevation of liver enzymes [41]. The median time to onset is around 10 weeks but can manifest as early as 2–3 weeks following the initiation of treatment. Fulminant liver failure can result but is rare. The underlying mechanism is not fully understood but may be multi-factorial, including the cross-reactivity of anti-tumor antibodies against hepatocytes, excessive cytokine production, and the development of autoantibodies [42]. Based on large clinical trials, ICIs are well tolerated among patients with underlying liver disease and the rate of ICH is comparable to that of patients without liver disease. However, these trials included only patients with Child–Pugh class A and B (7–9) cirrhosis [9,37,43].

4. Downstaging HCC with Neoadjuvant Immune Checkpoint Inhibitors

The use of ICIs as bridging or downstaging therapies prior to liver transplant has been described, although experience remains quite limited. A major concern limiting ICI use is the potential to increase the risk of acute rejection and lead to graft loss. In the post-transplant setting, studies on ICI use have shown that a significant portion of transplant recipients may experience biopsy-proven acute graft rejection when receiving ICIs for various malignancies. A recent review by Yin et al. [44] summarized outcomes on 28 liver transplant recipients who received ICIs. Eighteen received ICIs for HCC recurrence and the rest for melanoma and other types of cancers. Thirty-two percent of those patients experienced rejection. Similar rates of acute rejection were observed among kidney transplant recipients receiving ICIs, with over half of those that developed rejection losing their allograft [45].
To date, only six case reports have described ICI use either as a bridging or downstaging therapy for HCC prior to transplant [46,47,48,49,50,51]. All are single-center studies—two in the United States, two in Europe, and two in China. The 20 patients from these studies are presented in aggregate (Table 1). Most patients were male (85%) and had viral hepatitis as the cause of underlying liver disease. All except one received other forms of treatment, including surgical resection, sorafenib and LRT (information available for 13 patients only). Most patients (77%) were within the Milan criteria at the time of transplantation, whereas the rest were within the UCSF criteria. A range of ICIs were used as monotherapies, but all targeted the PD-1/PD-L1 pathway. Fifty-five percent of patients received nivolumab [46,47,51], whereas the rest received pembrolizumab, camrelizumab, toripalimab, or durvalumab [48,49,50]. The duration of ICI treatment varied from 6 weeks to over 2 years. The washout period between the last dose of ICI and liver transplant ranged from 1 day to 253 days.
Outcomes varied significantly among studies. Four studies reported successful transplant after ICI use [47,49,50,51]. Two patients (out of 18) had mild rejection that was successfully treated with the adjustment of their immunosuppression regimens. Poor outcomes were described in two studies [46,48], with both patients dying from liver failure early in the post-op course. Nordness et al. [46] described a patient with treated HCV who developed HCC recurrence following initial liver resection. His tumor burden was within the Milan criteria following sorafenib and Y90 treatment; however, he was not listed due to elevated AFP. Nivolumab was started and promptly controlled his disease progression. After a year without disease progression and remaining within the Milan criteria, he was listed for liver transplantation. Nivolumab was continued as a bridging therapy and the patient received a liver transplant from an HCV-antibody-positive NAT-negative donor, 8 days after his last dose of nivolumab. A donor liver biopsy showed no evidence of hepatitis, fibrosis, necrosis, or other abnormalities. Postoperatively, the liver enzymes failed to normalize, despite therapeutic tacrolimus levels and good vascular flow demonstrated on Doppler ultrasound and contrasted CT imaging. A biopsy on postoperative day 6 showed acute hepatic necrosis with dense lymphocyte infiltration. Despite salvage high-dose steroids and rabbit anti-thymocyte globulin (ATG) treatment, the patient continued to deteriorate and expired on day 10. Chen et al. [48] reported a similar case. Following recurrence after initial surgical resection, the patient received lenvatinib and toripalimab as a bridging therapy. The last dose of toripalimab was 93 days prior to the liver transplant. Despite an uncomplicated surgery, the patient did not regain liver function and eventually progressed to multi-organ failure and death on postoperative day 3. Liver biopsy on postoperative day 2 indicated massive hepatic necrosis. It is unclear whether ICI use was the culprit in this case, as there was no obvious evidence of rejection on pathology. The authors agreed that this could have represented primary graft nonfunction.
The washout period between the last dose of ICI and liver transplantation is clearly an important factor to consider. Schwacha-Eipper et al. discontinued nivolumab for 6 weeks before activating the patient on the waitlist [47]. A similar strategy was employed by Sogbe et al., (3 months) [50]. However, there is insufficient evidence to support a minimal washout period. In their nine-patient cohort, Tabrizian et al. reported successful transplantations 1 day and 2 days after the last nivolumab infusion [51]. It is important to note that both patients had significant blood loss intraoperatively, prompting 30-unit and 15-unit red blood cell transfusions, respectively. The blood product replacement potentially accelerated the washout of ICIs. Nevertheless, cases of successful transplant within 4 weeks of their last ICI dose have been described [49,51]. Additionally, the minimal washout time was often set loosely based on the reported serum half-life for ICIs (Table 2); however, the occupancy of the ICI pharmacological target can remain high for significantly longer. For example, the serum half-life for nivolumab is 12–20 days, but a sustained occupancy of over 70% of PD-1 molecules on circulating T is are observed more than 2 months following a single infusion. In some patients, PD-1 occupancy can remain above 50% for 200 days after the last dose following repeated infusion [52,53]. This prolonged effect of ICIs is supported by the often-delayed onset of side effects among patients receiving ICIs [54]. Overall, it is possible that a short interval between ICI infusion and transplant increases the risk of acute rejection post-operatively, but the quality of evidence has been limited thus far. Serum half-life of ICIs may be a poor indicator when deciding the minimal washout period, given the long-lasting effect on target receptors. Furthermore, since ICI binding to their corresponding target is reversible, it may be worthwhile to consider modalities such as plasmapheresis in order to accelerate washout if needed. A minimal washout period, however, could be useful in a different context, to potentially stratify patients. Those who progress early, especially systemically, upon discontinuation of ICIs, may not derive a benefit from a liver transplant.
The PD-1/PD-L1 pathway is a key negative regulator of the immune response and is involved in organ acceptance after transplant [65]. Studies on transplant recipients receiving ICIs suggest a correlation of graft PD-L1 expression with risk of graft rejection following ICI [66,67,68]. Of patients whose graft biopsy stained negative for PD-L1, none experienced acute rejection following nivolumab or pembrolizumab infusion, whereas those whose allograft stained positive for PD-L1 all experienced acute rejection [67,68]. Therefore, PD-L1 expression on liver allografts could theoretically serve as a potential marker in the context of anti-PD-1 ICI use. Its utility during the peri-transplant period, however, remains unknown. Both Nordness et al. and Chen et al. reported graft PD-L1 expression on immunohistochemistry—negative staining on pre-implant biopsy but positive on post-operative biopsies [46,47,48]. However, PD-L1 expression correlates with immune activation and could be a result of acute rejection [69]. Lastly, the use of anti-CTLA4 therapy in the neoadjuvant setting prior to liver transplantation has not been reported in the literature. Some case reports on ipilimumab use in the transplant patient population suggest that targeting CTLA4 may be associated with a lower risk of rejection [70,71].
Although not the primary focus of this review, neoadjuvant ICI has been utilized prior to definitive liver resection as well. The primary aim of neoadjuvant ICI in this context is to achieve downstaging followed by resection for those initially presenting with unresectable disease. However, this approach also has the potential to reduce the risk of recurrence for those presenting with initially resectable tumors. In the first scenario (downstaging of unresectable tumors), two case studies to date have described the neoadjuvant use of TACE with tislelizumab or camrelizumab in HCC patients before resection [72,73]. Chao et al. [72] reported on a patient with underlying cirrhosis presenting with a large tumor involving both right and middle hepatic veins, without extrahepatic metastasis. Following two rounds of TACE and tislelizumab, imaging showed a complete radiological response. The tumor was then resected safely and there was no disease recurrence at 6-month follow-up. Xin et al. [73] described two similar cases. The first patient presented with a large liver primary tumor, associated satellite lesions and a metastatic lesion in the spleen and the second patient presented with a bulky tumor with left and middle hepatic vein invasion and IVC tumor thrombosis. Both patients received TACE, neoadjuvant camrelizumab, and subsequent resection. Post-resection, both patients received adjuvant camrelizumab and had no recurrence on a 6-month follow-up. Given the small sample size and short follow-up, it remains unclear whether ICI should be utilized routinely before salvage resection. More clinical trials (NCT03510871 and NCT03299946) are currently in process to evaluate the efficacy of ICIs in borderline-resectable or unresectable HCC [74]. In the second scenario (the use of ICIs to reduce recurrence post-resection), early trial results have been encouraging, with a 37.5% pathological complete response observed in the interim analysis [75,76].

5. Conclusions

Liver transplant is a potentially curative option for patients who present with HCC beyond the Milan criteria. In additional to LRT, ICIs have recently emerged as a potential option for either bridging or downstaging therapy for those with intermediate- or advanced-stage HCC. The concern of an increasing risk of rejection associated with ICIs prior to transplant is valid; however, several case studies have demonstrated that this approach can be executed safely. A washout period between the last dose of ICIs and liver transplantation is ideal but may not be necessary, and a minimal washout period based on drug serum half-life can be problematic, especially since the PD-1 occupancy remains high even after 4–5 half-lives. Overall, the use of ICIs in a neoadjuvant setting is of significant interest to the field of HCC and transplant oncology. Clinical trials (NCT 04,035,876 and NCT 04425226) [77,78] are currently underway and will provide some insights on the safety profile and efficacy of ICI use in this setting.

Author Contributions

Conceptualization, Q.G. and A.S.B.; methodology, Q.G. and I.J.A.; investigation, Q.G. and I.J.A.; writing—original draft preparation, Q.G., I.J.A. and N.A.; writing—review and editing, Q.G., I.J.A., N.A. and A.S.B.; supervision, A.S.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef]
  2. Beumer, B.R.; de Wilde, R.F.; Metselaar, H.J.; de Man, R.A.; Polak, W.G.; Ijzermans, J.N.M. The Treatment Effect of Liver Transplantation versus Liver Resection for HCC: A Review and Future Perspectives. Cancers 2021, 13, 3730. [Google Scholar] [CrossRef] [PubMed]
  3. Mazzaferro, V.; Regalia, E.; Doci, R.; Andreola, S.; Pulvirenti, A.; Bozzetti, F.; Montalto, F.; Ammatuna, M.; Morabito, A.; Gennari, L. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N. Engl. J. Med. 1996, 334, 693–699. [Google Scholar] [CrossRef]
  4. Kardashian, A.; Florman, S.S.; Haydel, B.; Ruiz, R.M.; Klintmalm, G.B.; Lee, D.D.; Taner, C.B.; Aucejo, F.; Tevar, A.D.; Humar, A.; et al. Liver Transplantation Outcomes in a U.S. Multicenter Cohort of 789 Patients with Hepatocellular Carcinoma Presenting beyond Milan Criteria. Hepatology 2020, 72, 2014–2028. [Google Scholar] [CrossRef] [PubMed]
  5. Doyle, M.B.; Vachharajani, N.; Maynard, E.; Shenoy, S.; Anderson, C.; Wellen, J.R.; Lowell, J.A.; Chapman, W.C. Liver transplantation for hepatocellular carcinoma: Long-term results suggest excellent outcomes. J. Am. Coll. Surg. 2012, 215, 19–28, discussion 28–30. [Google Scholar] [CrossRef] [PubMed]
  6. Roayaie, S.; Obeidat, K.; Sposito, C.; Mariani, L.; Bhoori, S.; Pellegrinelli, A.; Labow, D.; Llovet, J.M.; Schwartz, M.; Mazzaferro, V. Resection of hepatocellular cancer ≤2 cm: Results from two Western centers. Hepatology 2013, 57, 1426–1435. [Google Scholar] [CrossRef]
  7. Franssen, B.; Jibara, G.; Tabrizian, P.; Schwartz, M.E.; Roayaie, S. Actual 10-year survival following hepatectomy for hepatocellular carcinoma. HPB 2014, 16, 830–835. [Google Scholar] [CrossRef] [Green Version]
  8. Llovet, J.M.; Ricci, S.; Mazzaferro, V.; Hilgard, P.; Gane, E.; Blanc, J.F.; de Oliveira, A.C.; Santoro, A.; Raoul, J.L.; Forner, A.; et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef]
  9. Finn, R.S.; Qin, S.; Ikeda, M.; Galle, P.R.; Ducreux, M.; Kim, T.Y.; Kudo, M.; Breder, V.; Merle, P.; Kaseb, A.O.; et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N. Engl. J. Med. 2020, 382, 1894–1905. [Google Scholar] [CrossRef]
  10. Sapisochin, G.; Hibi, T.; Toso, C.; Man, K.; Berenguer, M.; Heimbach, J.; Greten, T.F.; Pugh, T.J.; Dawson, L.A.; Mazzaferro, V. Transplant Oncology in Primary and Metastatic Liver Tumors: Principles, Evidence, and Opportunities. Ann. Surg. 2021, 273, 483–493. [Google Scholar] [CrossRef]
  11. Yao, F.Y.; Ferrell, L.; Bass, N.M.; Watson, J.J.; Bacchetti, P.; Venook, A.; Ascher, N.L.; Roberts, J.P. Liver transplantation for hepatocellular carcinoma: Expansion of the tumor size limits does not adversely impact survival. Hepatology 2001, 33, 1394–1403. [Google Scholar] [CrossRef]
  12. Yao, F.Y.; Xiao, L.; Bass, N.M.; Kerlan, R.; Ascher, N.L.; Roberts, J.P. Liver transplantation for hepatocellular carcinoma: Validation of the UCSF-expanded criteria based on preoperative imaging. Am. J. Transplant. 2007, 7, 2587–2596. [Google Scholar] [CrossRef] [PubMed]
  13. Mazzaferro, V.; Llovet, J.M.; Miceli, R.; Bhoori, S.; Schiavo, M.; Mariani, L.; Camerini, T.; Roayaie, S.; Schwartz, M.E.; Grazi, G.L.; et al. Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: A retrospective, exploratory analysis. Lancet Oncol. 2009, 10, 35–43. [Google Scholar] [CrossRef]
  14. Kaido, T.; Ogawa, K.; Mori, A.; Fujimoto, Y.; Ito, T.; Tomiyama, K.; Takada, Y.; Uemoto, S. Usefulness of the Kyoto criteria as expanded selection criteria for liver transplantation for hepatocellular carcinoma. Surgery 2013, 154, 1053–1060. [Google Scholar] [CrossRef] [PubMed]
  15. Toso, C.; Meeberg, G.; Hernandez-Alejandro, R.; Dufour, J.F.; Marotta, P.; Majno, P.; Kneteman, N.M. Total tumor volume and alpha-fetoprotein for selection of transplant candidates with hepatocellular carcinoma: A prospective validation. Hepatology 2015, 62, 158–165. [Google Scholar] [CrossRef]
  16. Sapisochin, G.; Goldaracena, N.; Laurence, J.M.; Dib, M.; Barbas, A.; Ghanekar, A.; Cleary, S.P.; Lilly, L.; Cattral, M.S.; Marquez, M.; et al. The extended Toronto criteria for liver transplantation in patients with hepatocellular carcinoma: A prospective validation study. Hepatology 2016, 64, 2077–2088. [Google Scholar] [CrossRef] [Green Version]
  17. Zheng, S.S.; Xu, X.; Wu, J.; Chen, J.; Wang, W.L.; Zhang, M.; Liang, T.-B.; Wu, L.-M. Liver transplantation for hepatocellular carcinoma: Hangzhou experiences: Retraction. Transplantation 2019, 103, 1736. [Google Scholar] [CrossRef]
  18. Nagai, S.; Kitajima, T.; Yeddula, S.; Salgia, R.; Schilke, R.; Abouljoud, M.S.; Moonka, D. Effect of Mandatory 6-Month Waiting Period on Waitlist and Transplant Outcomes in Patients with Hepatocellular Carcinoma. Hepatology 2020, 72, 2051–2062. [Google Scholar] [CrossRef]
  19. Kwong, A.J.; Kim, W.R.; Lake, J.R.; Smith, J.M.; Schladt, D.P.; Skeans, M.A.; Noreen, S.M.; Foutz, J.; Booker, S.E.; Cafarella, M.; et al. OPTN/SRTR 2019 Annual Data Report: Liver. Am. J. Transplant. 2021, 21 (Suppl. S2), 208–315. [Google Scholar] [CrossRef] [PubMed]
  20. Yao, F.Y.; Bass, N.M.; Nikolai, B.; Davern, T.J.; Kerlan, R.; Wu, V.; Ascher, N.L.; Roberts, J.P. Liver transplantation for hepatocellular carcinoma: Analysis of survival according to the intention-to-treat principle and dropout from the waiting list. Liver Transpl. 2002, 8, 873–883. [Google Scholar] [CrossRef]
  21. Coletta, M.; Nicolini, D.; Benedetti Cacciaguerra, A.; Mazzocato, S.; Rossi, R.; Vivarelli, M. Bridging patients with hepatocellular cancer waiting for liver transplant: All the patients are the same? Transl. Gastroenterol. Hepatol. 2017, 2, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Kulik, L.; Heimbach, J.K.; Zaiem, F.; Almasri, J.; Prokop, L.J.; Wang, Z.; Murad, M.H.; Mohammed, K. Therapies for patients with hepatocellular carcinoma awaiting liver transplantation: A systematic review and meta-analysis. Hepatology 2018, 67, 381–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Agopian, V.G.; Harlander-Locke, M.P.; Ruiz, R.M.; Klintmalm, G.B.; Senguttuvan, S.; Florman, S.S.; Haydel, B.; Hoteit, M.; Levine, M.H.; Lee, D.D.; et al. Impact of Pretransplant Bridging Locoregional Therapy for Patients with Hepatocellular Carcinoma within Milan Criteria Undergoing Liver Transplantation: Analysis of 3601 Patients from the US Multicenter HCC Transplant Consortium. Ann. Surg. 2017, 266, 525–535. [Google Scholar] [CrossRef] [PubMed]
  24. Cucchetti, A.; Cescon, M.; Bigonzi, E.; Piscaglia, F.; Golfieri, R.; Ercolani, G.; Cristina Morelli, M.; Ravaioli, M.; Daniele Pinna, A. Priority of candidates with hepatocellular carcinoma awaiting liver transplantation can be reduced after successful bridge therapy. Liver Transpl. 2011, 17, 1344–1354. [Google Scholar] [CrossRef]
  25. Heimbach, J.K.; Kulik, L.M.; Finn, R.S.; Sirlin, C.B.; Abecassis, M.M.; Roberts, L.R.; Zhu, A.X.; Murad, M.H.; Marrero, J.A. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology 2018, 67, 358–380. [Google Scholar] [CrossRef] [Green Version]
  26. Chapman, W.C.; Majella Doyle, M.B.; Stuart, J.E.; Vachharajani, N.; Crippin, J.S.; Anderson, C.D.; Lowell, J.A.; Shenoy, S.; Darcy, M.D.; Brown, D.B. Outcomes of Neoadjuvant Transarterial Chemoembolization to Downstage Hepatocellular Carcinoma before Liver Transplantation. Ann. Surg. 2008, 248, 617–625. [Google Scholar] [CrossRef]
  27. Lewandowski, R.J.; Kulik, L.M.; Riaz, A.; Senthilnathan, S.; Mulcahy, M.F.; Ryu, R.K.; Ibrahim, S.M.; Sato, K.T.; Baker, T.; Miller, F.H.; et al. A comparative analysis of transarterial downstaging for hepatocellular carcinoma: Chemoembolization versus radioembolization. Am. J. Transplant. 2009, 9, 1920–1928. [Google Scholar] [CrossRef]
  28. Yao, F.Y.; Mehta, N.; Flemming, J.; Dodge, J.; Hameed, B.; Fix, O.; Hirose, R.; Fidelman, N.; Kerlan, R.K., Jr.; Roberts, J.P. Downstaging of hepatocellular cancer before liver transplant: Long-term outcome compared to tumors within Milan criteria. Hepatology 2015, 61, 1968–1977. [Google Scholar] [CrossRef] [Green Version]
  29. Mehta, N.; Guy, J.; Frenette, C.T.; Dodge, J.L.; Osorio, R.W.; Minteer, W.B.; Roberts, J.P.; Yao, F.Y. Excellent Outcomes of Liver Transplantation following Down-Staging of Hepatocellular Carcinoma to within Milan Criteria: A Multicenter Study. Clin. Gastroenterol. Hepatol. 2018, 16, 955–964. [Google Scholar] [CrossRef]
  30. Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef] [Green Version]
  31. Mazzaferro, V.; Citterio, D.; Bhoori, S.; Bongini, M.; Miceli, R.; De Carlis, L.; Colledan, M.; Salizzoni, M.; Romagnoli, R.; Antonelli, B.; et al. Liver transplantation in hepatocellular carcinoma after tumour downstaging (XXL): A randomised, controlled, phase 2b/3 trial. Lancet Oncol. 2020, 21, 947–956. [Google Scholar] [CrossRef]
  32. Federico, P.; Petrillo, A.; Giordano, P.; Bosso, D.; Fabbrocini, A.; Ottaviano, M.; Rosanova, M.; Silvestri, A.; Tufo, A.; Cozzolino, A.; et al. Immune Checkpoint Inhibitors in Hepatocellular Carcinoma: Current Status and Novel Perspectives. Cancers 2020, 12, 3025. [Google Scholar] [CrossRef] [PubMed]
  33. Meyer, T.; Fox, R.; Ma, Y.T.; Ross, P.J.; James, M.W.; Sturgess, R.; Stubbs, C.; Stocken, D.D.; Wall, L.; Watkinson, A.; et al. Sorafenib in combination with transarterial chemoembolisation in patients with unresectable hepatocellular carcinoma (TACE 2): A randomised placebo-controlled, double-blind, phase 3 trial. Lancet Gastroenterol. Hepatol. 2017, 2, 565–575. [Google Scholar] [CrossRef] [Green Version]
  34. Lencioni, R.; Llovet, J.M.; Han, G.; Tak, W.Y.; Yang, J.; Guglielmi, A.; Paik, S.W.; Reig, M.; Kim, D.Y.; Chau, G.Y.; et al. Sorafenib or placebo plus TACE with doxorubicin-eluting beads for intermediate stage HCC: The SPACE trial. J. Hepatol. 2016, 64, 1090–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Montasser, A.; Beaufrère, A.; Cauchy, F.; Bouattour, M.; Soubrane, O.; Albuquerque, M.; Paradis, V. Transarterial chemoembolisation enhances programmed death-1 and programmed death-ligand 1 expression in hepatocellular carcinoma. Histopathology 2021, 79, 36–46. [Google Scholar] [CrossRef] [PubMed]
  36. Kudo, M.; Finn, R.S.; Qin, S.; Han, K.H.; Ikeda, K.; Piscaglia, F.; Baron, A.; Park, J.W.; Han, G.; Jassem, J.; et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: A randomised phase 3 non-inferiority trial. Lancet 2018, 391, 1163–1173. [Google Scholar] [CrossRef] [Green Version]
  37. El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.Y.; Choo, S.P.; Trojan, J.; Welling, T.H.R.; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef]
  38. Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. Lancet Oncol. 2018, 19, 940–952. [Google Scholar] [CrossRef]
  39. Yau, T.; Kang, Y.K.; Kim, T.Y.; El-Khoueiry, A.B.; Santoro, A.; Sangro, B.; Melero, I.; Kudo, M.; Hou, M.M.; Matilla, A.; et al. Efficacy and Safety of Nivolumab Plus Ipilimumab in Patients with Advanced Hepatocellular Carcinoma Previously Treated with Sorafenib: The CheckMate 040 Randomized Clinical Trial. JAMA Oncol 2020, 6, e204564. [Google Scholar] [CrossRef]
  40. Donisi, C.; Puzzoni, M.; Ziranu, P.; Lai, E.; Mariani, S.; Saba, G.; Impera, V.; Dubois, M.; Persano, M.; Migliari, M.; et al. Immune Checkpoint Inhibitors in the Treatment of HCC. Front. Oncol. 2020, 10, 601240. [Google Scholar] [CrossRef]
  41. Remash, D.; Prince, D.S.; McKenzie, C.; Strasser, S.I.; Kao, S.; Liu, K. Immune checkpoint inhibitor-related hepatotoxicity: A review. World J. Gastroenterol. 2021, 27, 5376–5391. [Google Scholar] [CrossRef]
  42. König, D.; Läubli, H. Mechanisms of Immune-Related Complications in Cancer Patients Treated with Immune Checkpoint Inhibitors. Pharmacology 2021, 106, 123–136. [Google Scholar] [CrossRef]
  43. Finn, R.S.; Ryoo, B.Y.; Merle, P.; Kudo, M.; Bouattour, M.; Lim, H.Y.; Breder, V.; Edeline, J.; Chao, Y.; Ogasawara, S.; et al. Pembrolizumab as Second-Line Therapy in Patients with Advanced Hepatocellular Carcinoma in KEYNOTE-240: A Randomized, Double-Blind, Phase III Trial. J. Clin. Oncol. 2020, 38, 193–202. [Google Scholar] [CrossRef]
  44. Yin, C.; Baba, T.; He, A.R.; Smith, C. Immune checkpoint inhibitors in liver transplant recipients—A review of current literature. Hepatoma Res. 2021, 7, 52. [Google Scholar] [CrossRef]
  45. Murakami, N.; Mulvaney, P.; Danesh, M.; Abudayyeh, A.; Diab, A.; Abdel-Wahab, N.; Abdelrahim, M.; Khairallah, P.; Shirazian, S.; Kukla, A.; et al. A multi-center study on safety and efficacy of immune checkpoint inhibitors in cancer patients with kidney transplant. Kidney Int. 2021, 100, 196–205. [Google Scholar] [CrossRef] [PubMed]
  46. Nordness, M.F.; Hamel, S.; Godfrey, C.M.; Shi, C.; Johnson, D.B.; Goff, L.W.; O’Dell, H.; Perri, R.E.; Alexopoulos, S.P. Fatal hepatic necrosis after nivolumab as a bridge to liver transplant for HCC: Are checkpoint inhibitors safe for the pretransplant patient? Am. J. Transplant. 2020, 20, 879–883. [Google Scholar] [CrossRef] [PubMed]
  47. Schwacha-Eipper, B.; Minciuna, I.; Banz, V.; Dufour, J.F. Immunotherapy as a Downstaging Therapy for Liver Transplantation. Hepatology 2020, 72, 1488–1490. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, G.-H.; Wang, G.-B.; Huang, F.; Qin, R.; Yu, X.-J.; Wu, R.-L.; Hou, L.-J.; Ye, Z.-H.; Zhang, X.-H.; Zhao, H.-C. Pretransplant use of toripalimab for hepatocellular carcinoma resulting in fatal acute hepatic necrosis in the immediate postoperative period. Transpl. Immunol. 2021, 66, 101386. [Google Scholar] [CrossRef]
  49. Qiao, Z.Y.; Zhang, Z.J.; Lv, Z.C.; Tong, H.; Xi, Z.F.; Wu, H.X.; Chen, X.S.; Xia, L.; Feng, H.; Zhang, J.J.; et al. Neoadjuvant Programmed Cell Death 1 (PD-1) Inhibitor Treatment in Patients with Hepatocellular Carcinoma before Liver Transplant: A Cohort Study and Literature Review. Front. Immunol. 2021, 12, 653437. [Google Scholar] [CrossRef]
  50. Sogbe, M.; López-Guerra, D.; Blanco-Fernández, G.; Sangro, B.; Narváez-Rodriguez, I. Durvalumab as a Successful Downstaging Therapy for Liver Transplantation in Hepatocellular Carcinoma: The Importance of a Wash-Out Period. Transplantation 2021, 105, e398–e400. [Google Scholar] [CrossRef] [PubMed]
  51. Tabrizian, P.; Florman, S.S.; Schwartz, M.E. PD-1 inhibitor as bridge therapy to liver transplantation? Am. J. Transplant. 2021, 21, 1979–1980. [Google Scholar] [CrossRef]
  52. Brahmer, J.R.; Drake, C.G.; Wollner, I.; Powderly, J.D.; Picus, J.; Sharfman, W.H.; Stankevich, E.; Pons, A.; Salay, T.M.; McMiller, T.L.; et al. Phase I Study of Single-Agent Anti–Programmed Death-1 (MDX-1106) in Refractory Solid Tumors: Safety, Clinical Activity, Pharmacodynamics, and Immunologic Correlates. J. Clin. Oncol. 2010, 28, 3167–3175. [Google Scholar] [CrossRef] [PubMed]
  53. Centanni, M.; Moes, D.J.A.R.; Trocóniz, I.F.; Ciccolini, J.; van Hasselt, J.G.C. Clinical Pharmacokinetics and Pharmacodynamics of Immune Checkpoint Inhibitors. Clin. Pharmacokinet. 2019, 58, 835–857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Spain, L.; Diem, S.; Larkin, J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat. Rev. 2016, 44, 51–60. [Google Scholar] [CrossRef] [PubMed]
  55. Feng, Y.; Masson, E.; Dai, D.; Parker, S.M.; Berman, D.; Roy, A. Model-based clinical pharmacology profiling of ipilimumab in patients with advanced melanoma. Br. J. Clin. Pharmacol. 2014, 78, 106–117. [Google Scholar] [CrossRef]
  56. Camacho, L.H.; Antonia, S.; Sosman, J.; Kirkwood, J.M.; Gajewski, T.F.; Redman, B.; Pavlov, D.; Bulanhagui, C.; Bozon, V.A.; Gomez-Navarro, J.; et al. Phase I/II Trial of Tremelimumab in Patients with Metastatic Melanoma. J. Clin. Oncol. 2009, 27, 1075–1081. [Google Scholar] [CrossRef]
  57. Bajaj, G.; Wang, X.; Agrawal, S.; Gupta, M.; Roy, A.; Feng, Y. Model-Based Population Pharmacokinetic Analysis of Nivolumab in Patients with Solid Tumors. CPT Pharmacomet. Syst. Pharm. 2017, 6, 58–66. [Google Scholar] [CrossRef]
  58. Elassaiss-Schaap, J.; Rossenu, S.; Lindauer, A.; Kang, S.P.; de Greef, R.; Sachs, J.R.; de Alwis, D.P. Using Model-Based “Learn and Confirm” to Reveal the Pharmacokinetics-Pharmacodynamics Relationship of Pembrolizumab in the KEYNOTE-001 Trial. CPT Pharmacomet. Syst. Pharm. 2017, 6, 21–28. [Google Scholar] [CrossRef]
  59. Papadopoulos, K.P.; Johnson, M.L.; Lockhart, A.C.; Moore, K.; Falchook, G.S.; Formenti, S.C.; Naing, A.; Carvajal, R.D.; Rosen, L.S.; Weiss, G.J.; et al. First-In-Human Study of Cemiplimab Alone or In Combination with Radiotherapy and/or Low-dose Cyclophosphamide in Patients with Advanced Malignancies. Clin. Cancer Res. 2020, 26, 1025–1033. [Google Scholar] [CrossRef] [Green Version]
  60. Yang, J.; Dong, L.; Yang, S.; Han, X.; Han, Y.; Jiang, S.; Yao, J.; Zhang, Z.; Zhang, S.; Liu, P.; et al. Safety and clinical efficacy of toripalimab, a PD-1 mAb, in patients with advanced or recurrent malignancies in a phase I study. Eur. J. Cancer 2020, 130, 182–192. [Google Scholar] [CrossRef]
  61. Zhang, L.; Mai, W.; Jiang, W.; Geng, Q. Sintilimab: A Promising Anti-Tumor PD-1 Antibody. Front. Oncol. 2020, 10, 594558. [Google Scholar] [CrossRef] [PubMed]
  62. Heery, C.R.; O’Sullivan-Coyne, G.; Madan, R.A.; Cordes, L.; Rajan, A.; Rauckhorst, M.; Lamping, E.; Oyelakin, I.; Marté, J.L.; Lepone, L.M.; et al. Avelumab for metastatic or locally advanced previously treated solid tumours (JAVELIN Solid Tumor): A phase 1a, multicohort, dose-escalation trial. Lancet. Oncol. 2017, 18, 587–598. [Google Scholar] [CrossRef]
  63. Baverel, P.G.; Dubois, V.F.S.; Jin, C.Y.; Zheng, Y.; Song, X.; Jin, X.; Mukhopadhyay, P.; Gupta, A.; Dennis, P.A.; Ben, Y.; et al. Population Pharmacokinetics of Durvalumab in Cancer Patients and Association with Longitudinal Biomarkers of Disease Status. Clin. Pharmacol. Ther. 2018, 103, 631–642. [Google Scholar] [CrossRef] [Green Version]
  64. Stroh, M.; Winter, H.; Marchand, M.; Claret, L.; Eppler, S.; Ruppel, J.; Abidoye, O.; Teng, S.L.; Lin, W.T.; Dayog, S.; et al. Clinical Pharmacokinetics and Pharmacodynamics of Atezolizumab in Metastatic Urothelial Carcinoma. Clin. Pharmacol. Ther. 2017, 102, 305–312. [Google Scholar] [CrossRef]
  65. Morita, M.; Fujino, M.; Jiang, G.; Kitazawa, Y.; Xie, L.; Azuma, M.; Yagita, H.; Nagao, S.; Sugioka, A.; Kurosawa, Y.; et al. PD-1/B7-H1 interaction contribute to the spontaneous acceptance of mouse liver allograft. Am. J. Transplant. 2010, 10, 40–46. [Google Scholar] [CrossRef] [PubMed]
  66. Friend, B.D.; Venick, R.S.; McDiarmid, S.V.; Zhou, X.; Naini, B.; Wang, H.; Farmer, D.G.; Busuttil, R.W.; Federman, N. Fatal orthotopic liver transplant organ rejection induced by a checkpoint inhibitor in two patients with refractory, metastatic hepatocellular carcinoma. Pediatr. Blood Cancer 2017, 64, e26682. [Google Scholar] [CrossRef] [PubMed]
  67. DeLeon, T.T.; Salomao, M.A.; Aqel, B.A.; Sonbol, M.B.; Yokoda, R.T.; Ali, A.H.; Moss, A.A.; Mathur, A.K.; Chascsa, D.M.; Rakela, J.; et al. Pilot evaluation of PD-1 inhibition in metastatic cancer patients with a history of liver transplantation: The Mayo Clinic experience. J. Gastrointest. Oncol. 2018, 9, 1054–1062. [Google Scholar] [CrossRef]
  68. Shi, G.M.; Wang, J.; Huang, X.W.; Huang, X.Y.; He, Y.F.; Ji, Y.; Chen, Y.; Wu, D.; Lu, J.C.; Sun, Q.M.; et al. Graft Programmed Death Ligand 1 Expression as a Marker for Transplant Rejection following Anti-Programmed Death 1 Immunotherapy for Recurrent Liver Tumors. Liver Transpl. 2021, 27, 444–449. [Google Scholar] [CrossRef]
  69. Choudhary, A.; Meijers, W.; Besharati, S.; Zhu, Q.; Lindenfeld, J.; Brinkley, M.; Anders, R.; Moslehi, J. Role of PD-L1 in heart transplant rejection. Eur. Heart J. 2020, 41, 1106. [Google Scholar] [CrossRef]
  70. Kittai, A.S.; Oldham, H.; Cetnar, J.; Taylor, M. Immune Checkpoint Inhibitors in Organ Transplant Patients. J. Immunother. 2017, 40, 277–281. [Google Scholar] [CrossRef]
  71. Fisher, J.; Zeitouni, N.; Fan, W.; Samie, F.H. Immune checkpoint inhibitor therapy in solid organ transplant recipients: A patient-centered systematic review. J. Am. Acad. Dermatol. 2020, 82, 1490–1500. [Google Scholar] [CrossRef]
  72. Chao, J.; Zhu, Q.; Chen, D.; An, X.; Liu, A.; Zhou, F.; Yuan, L.; Wang, Z.; Sun, H. Case Report: Transarterial Chemoembolization in Combination with Tislelizumab Downstages Unresectable Hepatocellular Carcinoma Followed by Radical Salvage Resection. Front. Oncol. 2021, 11, 667555. [Google Scholar] [CrossRef] [PubMed]
  73. Xin, H.; Zhang, C.; Ding, Z.; Zhang, M.; Ding, G.; Li, N. TACE plus PD-1 inhibitor (Camrelizumab) treatment for bridging to tumor resection in HCC: Case reports. Clin. Res. Hepatol. Gastroenterol. 2021, 46, 101777. [Google Scholar] [CrossRef]
  74. Yarchoan, M.; Zhu, Q.; Durham, J.N.; Gross, N.; Charmsaz, S.; Leatherman, J.M.; Zhang, S.; Popovic, A.; Weiss, M.J.; Philosophe, B.; et al. Feasibility and efficacy of neoadjuvant cabozantinib and nivolumab in patients with borderline resectable or locally advanced hepatocellular carcinoma (HCC). J. Clin. Oncol. 2021, 39, 335. [Google Scholar] [CrossRef]
  75. Kaseb, A.O.; Pestana, R.C.; Vence, L.M.; Blando, J.M.; Singh, S.; Ikoma, N.; Vauthey, J.N.; Allison, J.P.; Sharma, P. Randomized, open-label, perioperative phase II study evaluating nivolumab alone versus nivolumab plus ipilimumab in patients with resectable HCC. J. Clin. Oncol. 2019, 37, v880. [Google Scholar] [CrossRef]
  76. Pinato, D.J.; Cortellini, A.; Sukumaran, A.; Cole, T.; Pai, M.; Habib, N.; Spalding, D.; Sodergren, M.H.; Martinez, M.; Dhillon, T.; et al. PRIME-HCC: Phase Ib study of neoadjuvant ipilimumab and nivolumab prior to liver resection for hepatocellular carcinoma. BMC Cancer 2021, 21, 301. [Google Scholar] [CrossRef] [PubMed]
  77. 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] [PubMed]
  78. Bonney, G.K.; Chew, C.A.; Lodge, P.; Hubbard, J.; Halazun, K.J.; Trunecka, P.; Muiesan, P.; Mirza, D.F.; Isaac, J.; Laing, R.W.; et al. Liver transplantation for non-resectable colorectal liver metastases: The International Hepato-Pancreato-Biliary Association consensus guidelines. Lancet Gastroenterol. Hepatol. 2021, 6, 933–946. [Google Scholar] [CrossRef]
Table 1. Summary of characteristics and outcomes in patients who received ICIs as bridging and/or downstaging therapy prior to liver transplant.
Table 1. Summary of characteristics and outcomes in patients who received ICIs as bridging and/or downstaging therapy prior to liver transplant.
AuthorsAge/SexUnderlying Liver DiseaseMilan CriteriaICIDuration of ICIWashout Period AllowedDonor CharacteristicsDuration of Follow-UpComplicationRejectionRecurrence
Nordness, M.F. et al., 2020. Am J Transplant. [46]65/MHCVWithinNivolumab~2 years8 daysHCV antibody-positive, NAT-negativeDeath POD 10Hepatic necrosisN/AN/A
Schwacha-Eipper, B. et al., 2020. Hapatology. [47]62/MAlcoholic liver cirrhosisWithinNivolumab34 cycles15 weeksN/A1 yearNoneNoneNone
Chen, G.H. et al., 2021. Transplant Immunology. [48]39/MHBVWithinToripalimab10 cycles93 daysN/ADeath POD 3Hepatic necrosisN/AN/A
Sogbe, M. et al., 2021. Transplantation. [50]61/MHBVWithinDurvalumab15 months>90 daysN/A2 yearsNoneNoneNone
Tabrizian, P. et al., 2021. Am J Transplant. [51]69/MNonebeyond, within UCSF criteriaNivolumab21 cycles18 daysLiving donor23 monthsNoneNoneNone
56/FHCVbeyond, within UCSF criteriaNivolumab8 cycles22 daysN/A22 monthsNoneNoneNone
58/MHBVWithinNivolumab32 cycles1 days30 U intra-op transfusion22 monthsNoneNoneNone
63/MHCV, HIVWithinNivolumab4 cycles2 days15 U intra-op transfusion21 monthsNoneNoneNone
30/MHBVWithinNivolumab25 cycles22 daysN/A16 monthsNoneMild rejectionNone
63/MHBV, HIVWithinNivolumab4 cycles13 daysN/A14 monthsBile leakNoneNone
66/MHBVWithinNivolumab9 cycles253 daysN/A14 monthsNoneNoneNone
55/FHBVwithinNivolumab12 cycles7 daysN/A8 monthsNoneNoneNone
53/FNASHbeyond, within UCSF criteriaNivolumab2 cycles30 daysN/A8 monthsNoneNoneNone
Qiao, Z. et al., 2021. Front Immunol. [49]7 total. All male, age 53 ± 12.1N/AN/APembrolizumab or camrelizumabN/A1.3 months on averageN/AN/AN/A1/7 had mild acute rejectionN/A
N/A: not applicable. NAT: nucleic acid test. POD: postoperative day.
Table 2. Summary of major ICIs and their associated mechanisms of action and half-lives.
Table 2. Summary of major ICIs and their associated mechanisms of action and half-lives.
Immune Checkpoint InhibitorMechanism of ActionHalf-LifeReference
Ipilimumabanti CTLA-4 14.7 days [55]
Tremelimumabanti CTLA-4 19.6 days [56]
Nivolumabanti PD-125 days[57]
Pembrolizumabanti PD-114–27.3 days[58]
Cemiplimabanti PD-112 days[59]
Toripalimabanti PD-114.2 days[60]
Sintilimabanti PD-135.6 h[61]
Avelumabanti PD-L13.9–4.1 days[62]
Durvalumabanti PD-L121 days[63]
Atezolizumabanti PD-L127 days[64]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gao, Q.; Anwar, I.J.; Abraham, N.; Barbas, A.S. Liver Transplantation for Hepatocellular Carcinoma after Downstaging or Bridging Therapy with Immune Checkpoint Inhibitors. Cancers 2021, 13, 6307. https://doi.org/10.3390/cancers13246307

AMA Style

Gao Q, Anwar IJ, Abraham N, Barbas AS. Liver Transplantation for Hepatocellular Carcinoma after Downstaging or Bridging Therapy with Immune Checkpoint Inhibitors. Cancers. 2021; 13(24):6307. https://doi.org/10.3390/cancers13246307

Chicago/Turabian Style

Gao, Qimeng, Imran J. Anwar, Nader Abraham, and Andrew S. Barbas. 2021. "Liver Transplantation for Hepatocellular Carcinoma after Downstaging or Bridging Therapy with Immune Checkpoint Inhibitors" Cancers 13, no. 24: 6307. https://doi.org/10.3390/cancers13246307

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