Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jan 15, 2022; 14(1): 163-180
Published online Jan 15, 2022. doi: 10.4251/wjgo.v14.i1.163
Immunotherapy in liver transplantation for hepatocellular carcinoma: Pros and cons
Yi Luo, Fei Teng, Hong Fu, Guo-Shan Ding, Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
ORCID number: Yi Luo (0000-0003-1455-4162); Fei Teng (0000-0001-9076-8862); Hong Fu (0000-0003-0223-1905); Guo-Shan Ding (0000-0001-8917-969X).
Author contributions: Luo Y and Teng F contributed to the conception and design of the study; Luo Y performed the literature review, data analysis, and manuscript drafting; Teng F made critical revisions and was responsible for the final manuscript; Fu H participated in data analysis; Ding GS was the supervisor and participated in the manuscript editing.
Supported by National Natural Science Foundation of China, No. 81702923 and No. 81871262.
Conflict-of-interest statement: The authors declare that they have no conflicts of interest to report.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Fei Teng, MD, PhD, Attending Doctor, Surgeon, Surgical Oncologist, Department of Liver Surgery and Organ Transplantation, Changzheng Hospital, Naval Medical University, No. 415 Fengyang Road, Huangpu District, Shanghai 200003, China. tengfei@smmu.edu.cn
Received: March 22, 2021
Peer-review started: March 22, 2021
First decision: June 16, 2021
Revised: June 30, 2021
Accepted: December 7, 2021
Article in press: December 7, 2021
Published online: January 15, 2022

Abstract

Liver transplantation (LT) has emerged as a curative strategy for hepatocellular carcinoma (HCC), but contributes to a higher predisposition to HCC recurrence in the immunosuppression context, especially for tumors beyond the Milan criteria. Although immunotherapy has dramatically improved survival for immunocompetent patients and has become the standard of care for a variety of tumors, including HCC, it is mainly used outside the scope of organ transplantation owing to potentially fatal allograft rejection. Nevertheless, accumulative evidence has expanded the therapeutic paradigms of immunotherapy for HCC, from downstaging or bridging management in the pretransplant setting to the salvage or adjuvant strategy in the posttransplant setting. Generally, immunotherapy mainly includes immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT) and vaccine therapy. ICIs, followed by ACT, have been most investigated in LT, with some promising results. Because of the complex tumor microenvironment and immunoreactivity when immunosuppressants are combined with immunotherapy, it is difficult to reach formulations for immunosuppressant adjustment and the optimal selection of immunotherapy as well as patients. In addition, the absence of effective biomarkers for identifying rejection and tumor response is still an unresolved barrier to successful clinical immunotherapy applications for LT. In this review, we comprehensively summarize the available evidence of immunotherapy used in LT that is specific to HCC. Moreover, we discuss clinically concerning issues regarding the concurrent goals of graft protection and antitumor response.

Key Words: Hepatocellular carcinoma, Liver transplantation, Immunotherapy, Immune checkpoint inhibitors, Adoptive cell transfer, Immunosuppressant

Core Tip: This review addresses revolutionized immunotherapy for hepatocellular carcinoma (HCC) in liver transplantation (LT), from downstaging or bridging management in the pretransplant setting to adjuvant or salvage strategy in the posttransplant setting. Considering that the benefit of the antitumor response outweighs the incremental risk of rejection, it is worthwhile to take immunotherapy into account as the salvage option when HCC recurs after LT. More prospective studies are required to provide direct evidence regarding immunosuppressant adjustment, biomarkers for response and the optimal selection of immunotherapy as well as patients.



INTRODUCTION

Hepatocellular carcinoma (HCC) accounts for the majority of primary liver cancers, is the fourth leading cause of cancer-related death and is the sixth most commonly diagnosed cancer worldwide[1]. Liver transplantation (LT) is a well-established and highly effective curative therapy for HCC patients with limited tumor burden who are not candidates for resection. However, even for those that meet the strictest Milan criteria based on the explant tumor burden (i.e., a single nodule ≤ 5 cm in diameter or up to three nodules, with none larger than 3 cm in diameter and without tumor invasion into blood vessels or lymph nodes), the risk of HCC recurrence at 5 years after LT is estimated to be 10% to 15%[2]. Moreover, many countries outside the United States adopt expanded criteria rather than the Milan criteria, leading to an even higher incidence of HCC recurrence. When HCC recurs, the fate of the liver transplant recipient may be worse than that of the inoperable patient with advanced HCC, as immune checkpoint inhibitors (ICIs), the most significant breakthrough in recent years in cancer immunotherapy, are used outside the scope of transplantation. Immunotherapy has dramatically improved the survival of immunocompetent patients, with a long-term response and even complete cancer remission, and has become the standard of care for a variety of tumors, including HCC[3]. Immunotherapy, either by reactivating the suppressed intrinsic immune response or by transferring engineered immune cells, is aimed at immunopotentiation to eliminate tumors, which is contrary to immunosuppression for graft protection after transplantation. Therefore, rejection is an inherent risk for liver transplant recipients receiving immunotherapy and presents as a severe pattern that usually progresses rapidly to induce graft loss. In contrast, some patients receive immunotherapy without any sign of rejection, not only in LT but also in other solid organ transplantations. Generally, immunotherapy includes ICIs, adoptive cell transfer (ACT) and vaccine therapy[4]. Currently, most of the published studies on immunotherapy in the setting of LT are related to ICIs, followed by ACT, while vaccine therapy in LT has not been reported thus far. The different types of immunotherapies, as well as different immunosuppressants, have distinct mechanisms of action. When immunotherapy is combined with immunosuppressants in the setting of transplant recipients with malignancies, the interaction among the immune system, graft and cancer is mediated by a much more complex network of biological pathways than any of these entities alone. Many questions regarding the efficacy and safety of immunotherapy in this subgroup of patients remain unanswered. A recent review analyzed 91 patients treated with ICIs after kidney, liver or heart transplantation for different types of cancer and showed that 37 (41%) experienced rejection. Eight (10%) of 80 patients with an available survival status died due to rejection of the transplant, and 41 (51%) died of cancer progression[5]. As cancer progression is a greater threat and because immunotherapy appears to be the last therapeutic option for these patients, it is worth the risk of rejection. In this review, we focus on immunotherapy that is specific to HCC and used perioperatively in liver transplant recipients. We also discuss clinically concerning issues regarding the concurrent goals of graft protection and antitumor response that warrant further investigation.

IMMUNOTHERAPY AS A DOWNSTAGING OR BRIDGING APPROACH TO LT FOR HCC PATIENTS

The American Association for the Study of Liver Diseases suggests that patients beyond the Milan criteria be considered for LT after successful downstaging into the Milan criteria, which has been accepted by the United Network for Organ Sharing and provides a means for making formerly ineligible patients eligible for transplantation. For a long time, ablation and transarterial therapies have been used as two main downstaging approaches as well as bridging approaches, reducing the drop-out risk in the waiting list. Currently, immunotherapy is joining this oncological armamentarium, as an increasing number of clinical trials have shown encouraging objective response rates, even a complete response rate as high as 5.5%[3,4]. To date, 11 reported cases have used immunotherapy before LT (Table 1): 9 in a single-center series and 2 in two separate reports[6-8]. All 11 patients were treated with nivolumab, a programmed cell death protein-1 (PD-1) monoclonal antibody that belongs to ICIs, at a dose of 240 mg every 2 wk. The intra- and posttransplant immunosuppressant regimens were similar. One patient developed acute hepatic necrosis on postoperative day 5 that was likely related to the preoperative use of nivolumab and refractory to high-dose methylprednisolone and rabbit antithymocyte globulin and died on postoperative day 10. Another patient developed acute rejection, probably due to low tacrolimus levels, and responded rapidly to increasing dosages. The native liver explants of 4 patients showed > 90% tumor necrosis. After a follow-up of 16.0 ± 5.8 mo, none of the 10 surviving patients developed tumor recurrence.

Table 1 Characteristics of hepatocellular carcinoma patients receiving immunotherapy as a downstaging or bridging approach to liver transplantation.
No.
Ref.
Age
Sex
Underlying liver disease
MTD (cm)
Pathology milan in/out
Cycles/duration
Immunotherapy
Days before LT
Post-LT follow-up (mo)
Initial immunosuppression
Rejection
1Tabrizian et al[6]69MNone10Milan out within UCSF21 cyclesNivolumab1823Tapering steroids + tacrolimus + MMFNo
2Tabrizian et al[6]56FHCV5.4Milan out within UCSF8 cyclesNivolumab2222Tapering steroids + tacrolimus + MMFNo
3Tabrizian et al[6]58MHBV21Milan in32 cyclesNivolumab122Tapering steroids + tacrolimus + MMFNo
4Tabrizian et al[6]63MHCV, HIV4.4Milan in4 cyclesNivolumab221Tapering steroids + tacrolimus + MMFNo
5Tabrizian et al[6]30MHBV3.2Milan in25 cyclesNivolumab2216Tapering steroids + tacrolimus + MMFMild
6Tabrizian et al[6]63MHBV2Milan in4 cyclesNivolumab1314Tapering steroids + tacrolimus + MMFNo
7Tabrizian et al[6]66MHBV2.5Milan in9 cyclesNivolumab25314Tapering steroids + tacrolimus + MMFNo
8Tabrizian et al[6]55FHBV2.8Milan in12 cyclesNivolumab78Tapering steroids + tacrolimus + MMFNo
9Tabrizian et al[6]53FNASH8.7Milan out within UCSF2 cyclesNivolumab308Tapering steroids + tacrolimus + MMFNo
10Schwacha-Eipper et al[7]66MAlcohol-associated liver cirrhosis6.4Milan out34 cyclesNivolumab10512NANo
11Nordness et al[8]65MHCV5.5Milan in2 yrNivolumab8Death at day 10Tacrolimus + MMF + steroidsYes

Due to the small sample size and selective bias, it was difficult to determine risk factors associated with fatal rejection for those receiving immunotherapy as a downstaging or bridging approach to LT. However, the patient with fatal hepatic necrosis provided some clues. First, he received the longest immunotherapy of nivolumab (nearly 2 years) and underwent LT shortly after the last dose (8 d before transplantation). However, it is worth noting that 3 other patients who received the last dose less than 8 d before LT did not experience rejection, and one even received the last dose 1 d before transplantation with a total duration of 64 wk. However, a short interval between the last dose and LT should be avoided, as the half-life period of nivolumab is approximately 4 wk. Second, pathology of his explant revealed complete tumor necrosis and no evidence of residual HCC. Currently, there are no guidelines proposed for when and how to discontinue or taper ICIs. However, when a patient receiving immunotherapy achieves stable or regressive disease and is listed as a potential candidate for LT, a taper strategy should be considered. Third, the donor of the patient was positive for the HCV antibody, although without active HCV viremia, and there was no evidence of hepatitis or fibrosis on back-table biopsy of the donor liver. The relationship between an HCV-positive donor liver and severe rejection in the setting of immunotherapy needs further investigation.

IMMUNOTHERAPY AS ADJUVANT THERAPY FOR HCC AFTER LT

LT completely removes the primary tumors as well as potential lesions within the diseased liver. Circulating tumor cells or extrahepatic undetected lesions are origins of HCC recurrence. Theoretically, adjuvant therapy after LT can eliminate residual tumor cells, as the tumor burden, if still present, decreases to the lowest level. However, current evidence does not support adjuvant systematic therapies with chemotherapy or sorafenib to reduce the risk of HCC recurrence after LT[9]. A retrospective cohort study of 60 HCC patients within the University of California San Francisco criteria, published in 2018, assessed the posttransplant antirecurrence efficacy of Licardin in single and multiple administrations, a radioisotope iodine (131I)-labeled antibody fragment targeting the HCC-associated antigen HAb18G/CD147, and showed that adjuvant therapy with Licardin significantly reduced HCC recurrence after LT and that multiple administrations had little additional antirecurrence efficacy[10]. However, subsequent studies with larger sample sizes are rare. Due to the unpredictable risk of rejection, which occurs mainly in transplant recipients taking ICIs, immunotherapy as adjuvant therapy after LT should be used cautiously. ACT using natural killer (NK) cells or cytokine-induced killer (CIK) cells seems to be safer than ICIs. Tanimine et al[11] reported adjuvant immunotherapy using liver allograft-derived NK cells in 24 HCC patients after living-donor LT at the 2015 American Transplant Congress and stated that the intravenous transfer of processed NK cells to recipients 4 d after LT with a median of 273.5 million cells/patient significantly improved the 5-year recurrence-free survival and overall survival rates of patients pathologically exceeding the Milan criteria without any safety issues. Another case report on adjuvant immunotherapy using 5 × 109 CIK cells for 4 cycles one month after LT also showed no severe adverse effects, including rejection[12]. If we can distinguish patients with a low risk of rejection, immunotherapy, especially with ICIs, will be a very promising adjuvant therapy for those at a high risk of HCC recurrence after LT because of its superior performance on tumor response compared with other systemic therapies.

IMMUNOTHERAPY FOR HCC RECURRENCE AFTER LT

As described previously, HCC patients after LT are exposed to an inevitable risk of HCC recurrence, and unfortunately, there is a limited therapeutic arsenal available for the HCC recurrence subpopulation with progressive disease (PD) after routine treatment failure. However, in more recent years, growing research on immunotherapeutic applications in the transplant setting has yielded promising results that have revolutionized the therapeutic landscape of cancer recurrence after transplantation. Thus far, the cumulative literature on transplant immunotherapy is primarily focused on kidney transplantation[5]. A multicenter retrospective study covering 69 kidney transplant patients receiving ICIs reported improved overall survival (OS) despite a concomitant increased risk of rejection[13]. Given the satisfactory clinical outcomes, mounting research has been conducted to explore the potential of immunotherapy in liver transplant recipients with recurrence or de novo malignancy. Various malignancies can occur after LT, and melanoma patients seem to exhibit a favorable tumor response to immunotherapy and acceptable rejection rate[14-16]. In a review of ICIs for 6 melanoma patients after LT, 2 achieved complete remission (CR), 2 achieved partial remission (PR), and the remaining 2 developed PD; of note, no patient experienced allograft rejection[14]. There are also emerging reports on HCC recurrence treated with immunotherapy after LT (Table 2). To our knowledge, 29 patients with HCC recurrence had received immunotherapy after LT: 19 received ICIs [PD-1 inhibitors in 15 patients and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) inhibitor in 4 patients], and 10 received cell-based immunotherapy [9 based on T cell receptor (TCR) T cells and one based on allogenic NK cells]. The median patient age was 56 (14-70) years, and 78% of patients were male. Among the patients with recurrence sites reported, patients who developed intrahepatic HCC recurrence alone after LT accounted for 11% (2/18), those who developed extrahepatic recurrence alone accounted for 56% (10/18), and those who developed both accounted for 33% (6/18). The estimation of the efficacy and safety of immunotherapy was performed based on the summarized data (Table 2).

Table 2 Characteristics and reported outcomes of published cases with hepatocellular carcinoma recurrence receiving immunotherapy after liver transplantation.
No.
Ref.
Age
Sex
HCC recurrence
Immunosuppression protocol before immunotherapy
Compound
Duration of IMT (wk)
Interval from LT to IMT (yr)
Graft rejection
Tumor respon-se
Follow-up (mo)
Cause of death
1De Toni and Gerbes[27]41MIR and ERLow-dose tacrolimusNivolumab301NoPD10-
2Friend et al[59]20MERSirolimusNivolumab44Yes, lethal (17 d)NA1OF (4 wk after ICI initiation)
3Friend et al[59]14MERTacrolimusNivolumab23Yes, lethal (7 d)NA1OF (5 wk after ICI initiation)
4Varkaris et al[25]70MERLow-dose tacrolimusPembrolizumab11.38NoPD3PD
5DeLeon et al[60]57MHCC recurrenceTacrolimusNivolumab5.12.7NoPD1.2Probably PD
6DeLeon et al[60]56MHCC recurrenceSirolimus + MMFNivolumab4.77.8NoPD1.1Probably PD
7DeLeon et al[60]35FHCC recurrenceTacrolimusNivolumab5.63.7NoPD1.3Probably PD
8DeLeon et al[60]64MHCC recurrenceTacrolimusNivolumab1.31.2NoNA0.3MOF
9DeLeon et al[60]68MHCC recurrenceSirolimusNivolumab3.91.1Yes (27 d)NA0.9PD
10Gassmann et al[58]53FEREverolimus + MMF + steroidsNivolumab23Yes, lethal (7 d)NA0.8OF (2 wk after ICI initiation)
11Rammohan et al[32]57MERTacrolimus + MMF + steroid + mTOR inhibitorPembrolizumab42.94.3NoCR10Alive
12Zhuang et al[90]54MERTacrolimusNivolumab622.7NoPD20PD
13Al Jarroudi et al[91]70MIRTacrolimusNivolumab8> 3.0Yes (45 d)NA4PD
14Al Jarroudi et al[91]62FERTacrolimusNivolumab102.5NoPD2.5Alive
15Al Jarroudi et al[91]66MIR and ERTacrolimusNivolumab12> 4.75NoPD3Alive
16Amjad et al[24]62FIR and ER-Nivolumab82.71.3NoCR20Alive
17Wang et al[92]48MERSirolimus + tacrolimusPembrolizumab31Yes (5 d)NA8Alive
18Qiu et al[93]54MIR and ERSirolimusCamrelizumab394.3NoPD11PD
19Tan et al[21]56MERTacrolimus + MMFHBV-TCR T cells521.1NoPR12Alive
20Tan et al[21]45MIR and ERSirolimusHBV-TCR T cells164.4NoPD3.7Alive
21Qasim et al[20]70MERTacrolimusHBV-TCR T cells8.611NoPD2PD
22Hafezi et al[19]--HCC recurrenceTacrolimus + sirolimusHBV-TCR T cells101.5----
23Hafezi et al[19]--HCC recurrenceTacrolimus + sirolimus + MMFHBV-TCR T cells41----
24Hafezi et al[19]--HCC recurrenceTacrolimus + sirolimusHBV-TCR T cells91.8----
25Hafezi et al[19]--HCC recurrenceTacrolimus + MMFHBV-TCR T cells40.4----
26Hafezi et al[19]--HCC recurrenceSirolimusHBV-TCR T cells40.5----
27Hafezi et al[19]--HCC recurrenceTacrolimus + sirolimusHBV-TCR T cells80.7----
28Xie et al[22]29MIR-NK cells12.91.5NoPR18Alive
29Pandey and Cohen[49]54FIR and ERTacrolimusIpilimumab55.77.5NoCR27Alive
Efficacy: The last chance for liver transplant recipients who develop HCC recurrence

Multiple treatments were used before the initiation of immunotherapy, including sorafenib (n = 14), regorafenib (n = 5), lenvatinib (n = 2), chemotherapy (n = 7), radiotherapy (n = 5), transarterial chemoembolization (n = 4), ablation (n = 3) and surgery (n = 1), and all failed to control the disease. Therefore, salvage immunotherapy has been increasingly utilized as the last option for such subpopulations. Excluding patients whose responses were not reported or could not be assessed because of rapid progression to death or immunotherapy discontinuance after rejection, a total of 16 (55.2%) patients were eligible for response evaluation. The overall response rate (ORR) (CR + PR) was 31.3% (5/16) [including 18.8% (3/16) with CR and 12.5% (2/16) with PR], although 68.8% (11/16) of patients failed to respond to immunotherapy. In the ICI subgroup, the ORR was 25% (3/12), which manifested a numerically improved antitumor response in transplant patients compared to that in nontransplant patients with advanced HCC, where the ORRs to nivolumab and pembrolizumab were 15% and 18.3%, respectively[17,18]. Such a difference was difficult to interpret in terms of clinical benefit due to the limited sample size and selection bias, so further studies are necessary to establish whether each individual immunotherapy agent plays a different role through a specific mechanism in the liver transplant setting. For a single immunotherapy agent, the tumor response rates of patients treated with nivolumab, pembrolizumab, and ipilimumab were 11% (1/9), 50% (1/2), and 100% (1/1), respectively. This is discordant with a previously reported review across multiple organ transplantations, where the tumor response rates were 31% (8/26), 48% (12/25), and 29% (4/14), respectively[5].

On the other hand, 9 patients, all from a consecutive cohort led by researchers in Singapore, were treated with HBV-specific TCR T cells[19-21]. Three patients were reported to have a response, one achieved PR with a follow-up of 1 year, and two had PD. Furthermore, HBV-specific TCR T cells were engineered by researchers using the electroporation technique to gain short-term immunosuppressant resistance, which would be very promising in the setting of LT[19]. Another patient who achieved PR was an isolated case with a follow-up of 18 mo; in this patient, allogenic NK cells combined with iodine-125 seed implantation were used[22]. Whether NK cell transfer plays a dominant role in this combined immune-radiotherapy should be investigated through further studies.

In general, considering that the promising antitumor response outweighs the incremental risk of rejection when immunotherapy is used as a non-first-line protocol for liver transplant recipients who develop HCC recurrence, it is worthwhile to take immunotherapy into account as the last salvage option.

Safety: Rejection can be fatal, while PD inevitably leads to death

Because of the advanced stage of HCC when immunotherapy was administered, PD, which rapidly led to death, was the most common response status (12/16). The median duration of immunotherapy was 8.6 wk (IQR 4, 23 wk) and was not long enough to fully expose other immunotherapy-related adverse effects (irAEs) apart from rejection, which might also be related to immunosuppressant usage. Four patients developed grade 1-2 transaminitis, two patients developed a biliary stricture that needed stent implantation, and one patient experienced chills, fatigue, and fever. In the ICI immunotherapy subgroups, survival status was determined for 19 patients, and 32% (6/19), including 5 receiving nivolumab and 1 receiving pembrolizumab, experienced rejection. Interestingly, patients who developed both intra- and extrahepatic recurrence appeared to have a lower predisposition to rejection than those who developed intra- or extrahepatic recurrence alone; the incidence of rejection was 0% (0/5), 100% (1/1), and 50% (4/8), respectively. Allograft rejection exhibited a tendency to occur shortly after immunotherapy initiation, at a median time of 12 d (range 5-45 d). No difference in the interval from LT to ICI initiation was detected between patients who did and did not experience rejection (P = 0.191). The mean interval was 2.5 ± 1.2 years for those who experienced rejection and 4.0 ± 2.5 years for those who did not. Although statistical significance was not achieved, perhaps partially due to limited data, patients with a short interval seemed to be at a higher risk of rejection than those with a long interval. After a median follow-up of 3 (0.3-27) months, 68% (13/19) of patients died, but only 23% (3/13) of deaths were attributed to immediate rejection. This result was consistent with the preexisting literature on immunotherapy across multiple solid organ transplantation, which demonstrated that rejection-specific mortality was far less frequent than cancer-specific mortality (23% vs 77% in our pooled analysis)[5]. In addition, the graft rejection rates of patients treated with nivolumab, pembrolizumab, and ipilimumab were 36% (5/14), 33% (1/3), and 0% (0/1), respectively. In a systematic review of ICIs for organ transplant patients with a variety of cancers published in 2020[23], among all transplant recipients, the graft rejection rates of patients treated with nivolumab, pembrolizumab, and ipilimumab were 54.2%, 44% and 23%, respectively, and among all liver transplant recipients, the graft rejection rates were 33%, 25% and 12.8%, respectively. This tendency is consistent with our pooled analysis, which indicates that PD-1 inhibitors contribute to a higher risk of graft rejection than CTLA-4 inhibitors. Of note, one patient who experienced two episodes of acute cellular rejection before immunotherapy did not experience rejection after immunotherapy, which revealed that a history of rejection might not be a contraindication for immunotherapy[24].

In the cell-based immunotherapy subgroups, 10 patients received immune cell infusion, and 4 had evaluable graft rejection information. Notably, all 4 patients were successfully infused without severe irAEs or allograft rejection at a median duration of 10.8 wk (range 8.6-52 wk), which suggests that ACT might be superior to ICIs in terms of safety profile. Additionally, intensified immunosuppressive regimens were not applied during the ACT infusion, and tacrolimus-based immunosuppressive regimens accounted for 80% (8/10). Minimal but therapeutic immunosuppressive protocols merit further exploration for ACT immunotherapy.

Taken together, these results suggest that although allograft rejection can be fatal, the relatively low risk of rejection-associated death warrants consideration of immunotherapy as an alternative strategy because disease progression inevitably leads to death.

HOW TO BALANCE GRAFT-PROTECTIVE IMMUNOSUPPRESSION AND ANTITUMOR IMMUNOPOTENTIATION

Lifelong immunosuppression is required for liver transplant recipients to maintain graft protection. However, immunosuppressants might exert adverse pressure on the antitumor efficacy of immunotherapy by dampening host immune capacity[25,26]. According to the currently available data, favorable immunological and oncological responses are still obtained, even noninferior to those in the nontransplant setting, which suggests an incompletely antagonistic relationship between immunosuppression and the antitumor efficacy of immunotherapy. Nevertheless, on the one hand, conventional immunosuppressant regimens for liver recipients receiving immunotherapy may lead to neither graft rejection nor significant antitumor efficacy[27]. On the other hand, the usage of immunotherapy recommended for nontransplant HCC patients might not be fully applicable for liver transplant recipients who develop HCC recurrence. Therefore, how to balance graft-protective immunosuppression and antitumor immunopotentiation remains a critical issue, and further comprehensive investigations are required to explore individual usage and mechanisms in the simultaneous utilization of immunosuppression and immunotherapy.

Adjustment of the immunosuppressant regimen

The immunosuppressive microenvironment plays an important role in immune tolerance and graft protection. Currently, the major immunosuppressants used for liver recipients include calcineurin inhibitors (CNIs), steroids, antimetabolites, and mammalian target of rapamycin (mTOR) inhibitors, which inhibit T cell activation by blocking signaling pathways (signal 1: Antigen presentation and recognition, HLA-TCR/CD3, signal 2: Costimulatory signaling, and signal 3 cytokine priming)[28]. The major clinical immunosuppressants target signals 1 and 3, while cancer immunotherapy targets signal 2[29]. CNIs, such as FK506, which targets signal 1, partially block IL-2 expression by disrupting the activation of nuclear factor of activated T cells[28,30]. Due to the unquestionable capacity of rejection reduction, CNIs are extensively used for the majority of liver transplant recipients[31]. In our pooled analysis, 70% (19/27) of patients were administered a tacrolimus-based immunosuppression protocol during immunotherapy, and 3 achieved a tumor response (2 CRs and 1 PR). Of concern is that low-dose tacrolimus, the minimal immunosuppression strategy, does not increase the burden of rejection and concomitantly avoids interference with the antitumor immune activity of immunotherapy[25,27,32]. Different from CNIs, mTOR inhibitors, including sirolimus and everolimus, block signal 3 of final T cell activation by inhibiting the cell cycle transition from G1 to S phase and thereby influence both the proliferation and activation of T lymphocytes[33,34]. Additionally, mTOR inhibitors have antitumor properties, and as a result, mTOR inhibitors are inclined to be used for liver transplant patients with HCC[35]. However, whether mTOR inhibitors play an essential biological role in graft protection and antitumor efficacy for liver transplant patients who develop HCC recurrence remains unclear. More recently, two studies tended to support the notion that mTOR inhibitors had the potential to uncouple the efficacy and rejection of ICIs in renal transplantation[13,36]. Compared to non-mTOR inhibitor subsets, the administration of mTOR inhibitors in renal transplant patients with malignancy presented a lower predisposition to rejection and simultaneously resulted in improved rejection-free graft survival and overall graft survival[13]. Apart from the aforementioned immunosuppressants, an increasing number of immunosuppressants appear to be associated with a low risk of rejection without affecting the ORR of the tumor to ICIs. Therefore, based on preliminary evidence, regimens combining mTOR inhibitors with low-dose tacrolimus may warrant consideration as an alternative strategy.

Moreover, whether additional steroids may antagonize the therapeutic profile of immunotherapy also remains controversial. Murakami et al[13] reported that steroids can diminish the effect of immunotherapy. Conversely, some studies of immunotherapy for organ transplant patients indicated that additional steroids may not exert a negative effect on the efficacy of immunotherapy and may even decrease the risk of irAEs[32,37]. A systematic review involving 39 allograft transplant patients treated with ICIs revealed that individual immunosuppressive regimens had different effects on allograft rejection and tumor response[38]. The allograft rejection rates with a single agent, including prednisone, mTOR inhibitors, or CNIs, and the combination regimen were 78% (7/9), 67% (2/3), 11% (1/9), and 29% (5/17), respectively. The tumor response rates to ICIs were 63% (5/8), 50% (1/2), 25% (2/8), and 50% (7/14)[38]. It is presumed that a single steroid regimen may be insufficient to prevent rejection, despite a satisfactory tumor response. Thus, steroids combined with other low-dose immunosuppressants, such as CNIs and mTOR inhibitors, may yield promising outcomes in specifically stratified subgroups. Nevertheless, there is no definitive conclusion on the respective contributions of immunosuppressants in HCC patients after LT in our pooled analysis due to the absence of supporting information. Taken together, these findings indicate that given the limitation and heterogeneity of the experimental data, the optimal immunosuppressant regimen cannot be determined, and a combined strategy of mTOR inhibitors and low-dose tacrolimus, with or without steroids, warrants further validation.

Choice of immunotherapy and whether use it in a modified manner

The antitumor efficacy and rejection risk of each individual immunotherapy are distinctly different, and the identification of specific patients and selection of a reasonable management plan based on the respective biological properties of each immunotherapy are urgent matters. The most clinically relevant inhibitory costimulatory pathways (signal 2) are the PD-1:PD-L1/PD-L2 and CTLA-4/B7 axes, which are considered to function at different phases of the T cell response. Both of these inhibitory pathways contribute to immune tolerance; in addition, the PD-1 axis is thought to be the most essential for graft tolerance primarily during the maintenance phase across the posttransplant process, while the CTLA-4/B7 axis functions during the induction phase[39-41]. Therefore, PD-1 inhibitors (nivolumab, pembrolizumab) are more likely to give rise to graft rejection than CTLA-4 inhibitors (ipilimumab), as delineated in our analysis and a previous review[23]. Given that the CTLA-4 axis functions during the induction phase of immune tolerance, some studies have reported that CTLA-4 blockade at the late stage resulted in a lower risk of rejection than that at the early stage[23,42,43]. From the scant evidence, CTLA-4 inhibitors (ipilimumab) are likely more appropriate than PD-1 inhibitors for patients at a high risk of rejection or with a remote LT history.

PD-L2, unlike PD-L1 (the major ligand for PD-1 in peripheral tissues), is more commonly expressed on monocytes and dendritic cells than on tumor cells, and both PD-L1 and PD-L2 are considered to play crucial roles in allograft tolerance[44]. Therefore, from the clinical perspective, PD-L1-specific blockade (preventing the binding of PD-L1 to PD-1) may contribute to a lower predisposition to allograft rejection than PD-1 blockade (preventing the interactions of PD-1 with PD-L1 and PD-L2), partially owing to the preserved biological effects of the PD1/PD-L2 axis in immune tolerance. However, the therapeutic differences in activity and toxicity between PD-1 inhibitors and PD-L2 inhibitors remain to be further evaluated.

To date, no solid conclusion has been drawn regarding whether a modified method is required for immunotherapy. All patients with available information in our analysis were administered ICI immunotherapy in accordance with the instructions. From the perspective of the dose-effect relationship, low-dose exposure to nivolumab (≥ 0.3 mg/kg) could competitively saturate peripheral receptor occupancy and contribute to comparable antitumor efficacy[45,46]. In particular, low- but therapeutic-dose immunotherapy may not only relatively reduce adverse events and financial burden but also not compromise efficacy. Further prospective investigations are needed to explore the precise dose-effect relationship of each individual agent in HCC patients undergoing LT.

Notably, given that the efficacy of ICIs usually appears within 3 mo after initiation[47] and that PD-1 receptor occupancy lasts up to 85 d[48], a markedly prolonged duration is inadvisable because of the increased risk of rejection. Nordness et al[8] reported a case in which a recipient who received nivolumab for 2 years prior to LT developed fatal rejection, but pathology of his explants revealed complete tumor necrosis and no evidence of residual HCC. In another published case report, a partial tumor response occurred after three doses of ipilimumab (3 mg/kg), and CR was eventually achieved following the fourth dose of a 3-wk schedule conversion to a 12-wk schedule; notably, a durable response of 27 mo was obtained after a 13-mo ipilimumab regimen[49]. In view of the above results, a tumor response may develop at a relatively early stage, and a prolonged duration of immunotherapy would lead to immunotherapy resistance or severe adverse events. As a result, a prolonged cycle interval and even withdrawal need to be taken into consideration after a definitely complete tumor response based on periodic evaluations and timely identification.

Currently, the exploited cell subgroups of ACT mainly include tumor-infiltrating lymphocytes (TILs), CIK cells, lymphokine-activated killer cells, NK cells, T cells, and genetically redirected cells. In several accomplished studies, chimeric antigen receptor (CAR) T cell immunotherapy targeting tumor-associated antigens (TAAs) showed strong antitumor capacities but also nonnegligible adverse events, such as cytokine release syndrome (CRS) and neurotoxicity, which limited its clinical applications in the liver transplant setting[50,51]. Unlike CAR-T cell therapy, CAR-NK cell therapy rarely elicits CRS or neurotoxicity; thus, CAR-NK cell therapy might be more suitable for translation into organ transplantation[52]. In our pooled data, CIK cells, NK cells, and HBV-TCR T cells were used in a liver transplant setting with promising clinical results. However, there are many unsolved problems regarding highly efficient production, dosing adjustment, and identification of tumor-specific antigens. Based on existing experiences, dose escalation and a relatively low-dose regimen might be favorable in the liver transplant setting. Considering the high heterogeneity of HCC, engineered cells with multiple targets and combined regimens represent new frontiers.

As mentioned above, there is still no study reporting vaccine therapy in the setting of LT. Even in a nontransplant setting, only a few trials of vaccine therapy targeting HCC-associated antigens have been performed, and none of them has provided clinically meaningful results. However, a strategy using neoantigens has emerged as a promising approach to develop cancer vaccines with intense tumor-specific nontoxic responses due to advancements in the field of high-throughput screening. The ability to predict highly immunogenic neoantigens with antitumor activity as vaccines using this approach has been shown in melanoma[53] and glioblastoma[54]. Although vaccines are traditionally considered a stand-alone therapy, there is a tendency to combine them with ICIs or ACT.

Surveillance and management of immunotherapy-related rejection

Immunotherapy-related rejection remains the major barrier to clinical immunotherapy promotion in HCC patients after LT. For liver transplant recipients receiving immunotherapy, the identification of rejection is easily confounded by immune-related hepatitis, a kind of irAE, which is characterized mainly by mild transaminitis (grades 1-2)[55]. Thus, caution is strongly warranted to distinguish immune-related hepatitis and rejection when apparent liver malfunction is detected. Compared with rejection, hepatitis occurs at a later stage following immunotherapy initiation (median time, 22 d vs 5-6 wk)[5,55] and rarely leads to fatal outcomes. Beyond this, immune-related hepatitis is more common in patients treated with CTLA-4 inhibitors[56], whereas allograft rejection is more frequently recorded in liver transplant patients treated with PD-1 inhibitors[23]. When a definite diagnosis cannot be made by virtue of the information above, graft biopsy should be performed and evaluated based on the Banff schema[57]. Generally, immune-related hepatitis is primarily characterized by acute lobular hepatitis, whereas allograft rejection is predominantly characterized by portal inflammation, bile duct damage, and endotheliitis[58]. Clinically, hepatitis and rejection do not seem to be completely distinct, and to some extent, they could be partially homologous. If a single liver biopsy presents both pathological features simultaneously, it is difficult to identify potential mutual interactions involved in disease progression; therefore, given the potential benefit for rejection control, further studies are required to explore the underlying relationship of hepatitis and rejection.

In particular, surveillance should focus on stratified populations who tend to be susceptible to rejection. Although no difference was detected in the interval from LT to ICI initiation between patients who did and did not experience rejection (2.5 ± 1.2 years vs 4.0 ± 2.5 years, P = 0.191), patients with a narrow interval from LT to immunotherapy initiation exhibited a tendency to have a higher risk of rejection. Moreover, in our analysis, rejection usually occurred shortly after immunotherapy initiation, at a median time of 12 d (range 5-45 d) or at a short cycle (range 1-4 cycles); therefore, more intensive surveillance is recommended during the early period after immunotherapy initiation. Of concern, PD-L1 expression on graft lymphocytes was reported to be strongly associated with rejection after ICI initiation[59,60]; however, Nordness et al[8] reported a case of rejection whose PD-L1 staining appeared to be negative before transplantation but positive after transplantation. It can be speculated that PD-L1 expression manifests as a secondary phenomenon following rejection, and therefore, liver biopsy should be performed routinely to validate its predictive efficacy.

Since allograft rejection largely appears to be life-threatening, effective preventive and therapeutic interventions are critically required in clinical practice. Evidence indicates that a cellular-mediated mechanism plays a key role in graft rejection, whereas an antibody-mediated mechanism is secondary only to the former[61,62]. In accordance with this evidence, all 3 evaluable patients enrolled in our analysis experienced cellular-mediated rejection, and 2 experienced both cellular- and antibody-mediated rejection. Typically, in liver transplant recipients who do not receive ICI treatment, approximately 75% of acute cell-mediated rejection can be mitigated with high-dose steroids[58,63]. Comparatively, in this population taking ICIs, only 29% of patients with allograft rejection were salvaged throughout the treatment course; most patients experienced graft failure[23]. This is consistent with our analysis, where only 2 of 6 (33%) recipients showed a response to steroids. Furthermore, dialysis is often used as an alternative option for rejection in renal transplant recipients, but whether it is feasible in liver transplant recipients remains unclear[38]. Some scholars recommend plasmapheresis as a viable alternative solution for immunotherapy-induced rejection. Although plasmapheresis is mainly thought to alleviate acute antibody-mediated rejection rather than cell-mediated rejection, it can substantially accelerate clearance from the circulation and thus mitigate immunotherapy-induced rejection[58,64]. In addition, antithymocyte globulin and infliximab were reported to be successfully used for acute rejection in liver transplant recipients, but further investigation is needed[65,66]. In summary, an in-depth collaboration involving the patient, surgeon, and oncologist is urgently necessary to identify individualized risk-benefit profiles because of the absence of highly effective therapeutic means available.

Immunotherapy combined with other treatments

To achieve a higher response rate, combination strategies based on immunotherapy might be a promising direction toward optimal antitumor efficacy in liver transplant recipients who develop HCC recurrence. Combination with conventional HCC therapies is the first option. Locoregional liver-directed therapies, such as ablation and transarterial therapies, exhibit the dual effects of robust tumor destruction to liberate substantial TAAs and strongly activate the immune response by priming tumor-specific T cells[67]. Such therapy-induced immunogenic modulation of tumors might amplify the antitumor efficacy of CD8+ effector T cells activated by ICIs[67].

In addition, molecularly targeted therapies with immunotherapies have become the standard of care for advanced HCC. The FDA, EMA and other regulatory agencies worldwide have approved the PD-L1 inhibitor atezolizumab plus vascular endothelial growth factor (VEGF) inhibitor for first-line therapy in HCC. Atezolizumab plus bevacizumab is now listed as the preferred regimen in first-line systemic therapies by National Comprehensive Cancer Network guidelines for HCC, replacing sorafenib and lenvatinib[68,69]. The combination with lenvatinib was associated with double the response rate compared with that observed with single-agent pembrolizumab, but this came at the cost of increased toxicity[70]. In addition, tyrosine kinase inhibitors (TKIs), such as sorafenib, regorafenib and lenvatinib, have been shown to have immune-associated antitumor capacity independent of anti-VEGFR mechanisms[71]. Accumulative studies have demonstrated that sorafenib can stimulate antitumor efficacy by strengthening CD4+ and CD8+ T cell function and infiltration and inhibiting T-reg cells[72-74]. In the liver transplant setting, it has been reported that an HCC patient following LT developed metastatic lung lesions and subsequently received sorafenib but experienced disease progression after 1 year. Then, pembrolizumab was added to sorafenib treatment, and ultimately, the patient achieved CR without allograft rejection[32], which indicated the crucial synergistic antitumor efficacy of the combination of PD-1 inhibitors with TKIs even though TKIs failed as a first-line treatment. Currently, a number of phase III clinical trials using a combination of molecularly targeted therapies and immunotherapies are being conducted. If one or more of them also show positive results, the choice of preferred treatment will depend substantially on patient characteristics, tolerability and toxicity profile, and the preferred strategy would offer concrete experience to draw upon for HCC patients in the LT setting.

Growing evidence indicates that the gut microbiota affects the liver microenvironment in allograft rejection and HCC development[75-77]. Recently, several human studies have suggested that increased microbial diversity exerts a profound effect on the response to PD-1 inhibitors, which might be mediated by increased intratumor CD8+ T cell infiltration[78-80]. However, which specific bacterial taxa contribute to an improved tumor response to PD-1 inhibitors remains an unsolved issue. Hence, fecal microbiota transplantation (FMT), which shifts the entire gut microbiota to patients, may be an alternative. In the liver transplant setting, PD-1 inhibitors in combination with FMT might substantially improve the tumor response and allograft rejection, but more prospective studies are required.

Biomarkers for the response to immunotherapy

Effective biomarkers for identifying potential responders to ICIs would allow physicians to select optimal candidates for immunotherapy. PD-L1 expression on tumor cells was reported to be associated with the tumor response to PD-1 inhibitors[81]; however, in contrast, the CHECKMATE-040 trial suggested that the tumor response occurred regardless of PD-L1 staining[82]. Thus, PD-L1 expression in tumor tissues does not seem sufficient as a single predictor to identify potential responders to PD-1 blockade. It is thought that immunotherapies, particularly ICIs, work in part by reactivating preexisting TILs. TILs are a class of lymphocytes in the tumor microenvironment that affect carcinogenesis and include CD8+ T cells, CD4+ T cells, tumor-associated macrophages (TAMs), tumor-associated neutrophils, myeloid-derived suppressor cells (MDSCs) and NK cells. An increased density of specific TIL phenotypes, particularly activated CD8+ TILs, is correlated with small tumor size, early TNM stage and better prognosis in HCC patients[83], and the CD8+ TIL density of responders was higher than that of nonresponders[84]. In addition, positive TILs in the tumor margin might be more associated with the tumor response than those in the tumor center[85,86]. In the tumor microenvironment, CD8+ TILs are exhausted or dysfunctional. The failure of CD8+ TILs to kill tumor cells involves signals from multiple cells, including MDSCs, Tregs, and TAMs. The interaction of PD-L1 with PD-1 on CD8+ TILs causes suppression and a decrease in their effector function, leading to decreased tumor cell death. Furthermore, the galectin-9 and T cell immunoglobulin and mucin-domain containing (TIM)-3 interaction on MDSCs and IL-10 secretion by Tregs have a similar effect[87]. Therefore, TILs and PD-L1 should be combined to guide the development of immunotherapies and predict their clinical responses in cancers. A recent study by DeLeon et al[60], covering 5 recipients with PD-L1 staining and 4 with TIL assessments, presumed that the combined expression of PD-L1 and TILs might be more reliable in liver transplant recipients. Additionally, the KEYNOTE-224 trial established a score involving both PD-L1-positive tumor cells and the immune cell ratio to the total number of viable tumor cells, with a positive score indicating a higher likelihood of tumor response[88]. In addition to the markers mentioned above, microsatellite instability, mismatch repair deficiency, and tumor mutational burden were thought to be potential biomarkers for predicting the response to ICIs; however, whether these biomarkers work well in the liver transplant setting requires further investigation. Some predictive biomarkers have been proposed to identify which patients are likely to benefit from CTLA-4 blockade; these include the absolute lymphocyte count and T cell activation marker-inducible costimulator[89]. However, to date, no biomarker has been validated in liver transplant recipients with CTLA-4 blockade. Herein, given the frustration with the inability to identify specific responder subsets, PD-1 inhibitors might be taken into consideration prior to CTLA-4 inhibitors to maximize tumor response. In addition, it is recommended that liver biopsy be conducted both pre- and postimmunotherapy together with a relevant biomarker quantitative assessment for a better stratification of HCC patients after LT.

CONCLUSION

Within the last decade, breakthroughs in immunotherapy have greatly expanded the treatment armamentarium for HCC. However, there is still an unlit corner for HCC patients awaiting LT or after LT due to the deep concern about lethal rejection induced by immunotherapy. On the one hand, there will be an increasing number of HCC patients after immunotherapy who are bridged or downstaged to be candidates for LT, as immunotherapy is now gradually becoming a part of routine or even preferred regimens for HCC systemic therapy. There are also many patients with HCC recurrence after LT who fail to respond to other therapies, and immunotherapy may be their last option. We must face the demand for immunotherapy in the setting of LT. On the other hand, the rejection rate, especially the lethal pattern, is higher than we can afford, and there are many unsolved problems when immunotherapy coexists with immunosuppressants in the setting of LT. Therefore, we need to explore immunotherapies in LT for HCC with caution regarding immunosuppressant adjustment, biomarkers for safety and efficacy, and selection strategies for different immunotherapies and patients.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Oncology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Li L S-Editor: Gao CC L-Editor: A P-Editor: Gao CC

References
1.  Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53206]  [Cited by in F6Publishing: 50811]  [Article Influence: 8468.5]  [Reference Citation Analysis (44)]
2.  Mehta N, Heimbach J, Harnois DM, Sapisochin G, Dodge JL, Lee D, Burns JM, Sanchez W, Greig PD, Grant DR, Roberts JP, Yao FY. Validation of a Risk Estimation of Tumor Recurrence After Transplant (RETREAT) Score for Hepatocellular Carcinoma Recurrence After Liver Transplant. JAMA Oncol. 2017;3:493-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 218]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
3.  Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim TY, Kudo M, Breder V, Merle P, Kaseb AO, Li D, Verret W, Xu DZ, Hernandez S, Liu J, Huang C, Mulla S, Wang Y, Lim HY, Zhu AX, Cheng AL; IMbrave150 Investigators. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med. 2020;382:1894-1905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2542]  [Cited by in F6Publishing: 3317]  [Article Influence: 829.3]  [Reference Citation Analysis (1)]
4.  Kole C, Charalampakis N, Tsakatikas S, Vailas M, Moris D, Gkotsis E, Kykalos S, Karamouzis MV, Schizas D. Immunotherapy for Hepatocellular Carcinoma: A 2021 Update. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 79]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
5.  Delyon J, Zuber J, Dorent R, Poujol-Robert A, Peraldi MN, Anglicheau D, Lebbe C. Immune Checkpoint Inhibitors in Transplantation-A Case Series and Comprehensive Review of Current Knowledge. Transplantation. 2021;105:67-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
6.  Tabrizian P, Florman SS, Schwartz ME. PD-1 inhibitor as bridge therapy to liver transplantation? Am J Transplant. 2021;21:1979-1980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 58]  [Article Influence: 19.3]  [Reference Citation Analysis (1)]
7.  Schwacha-Eipper B, Minciuna I, Banz V, Dufour JF. Immunotherapy as a Downstaging Therapy for Liver Transplantation. Hepatology. 2020;72:1488-1490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 50]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
8.  Nordness MF, Hamel S, Godfrey CM, Shi C, Johnson DB, Goff LW, O'Dell H, Perri RE, Alexopoulos SP. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 72]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
9.  Pelizzaro F, Gambato M, Gringeri E, Vitale A, Cillo U, Farinati F, Burra P, Russo FP. Management of Hepatocellular Carcinoma Recurrence after Liver Transplantation. Cancers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
10.  Li J, Qin L, Ding EC, Shen J, Li JP. [Efficacy and safety of licartin with repeated administration in treatment of HCC patients after liver transplantation]. Zhonghua Yi Xue Za Zhi. 2018;98:2645-2649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
11.  Tanimine N, Tanaka Y, Ishiyama K, Ohira M, Shimizu S, Yano T, Ohdan H. Adoptive Immunotherapy with Liver allograft-derived NK Cells Improves Recurrence-free Survival after Living-donor Liver Transplantation inPatients with Hepatocellular Carcinoma [abstract]. Am J Transplant. 2015;15 (suppl 3).  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Li R, Yan F, Liu L, Li H, Ren B, Hui Z, Ren X. Cytokine-induced killer cell therapy for the treatment of primary hepatocellular carcinoma subsequent to liver transplantation: A case report. Oncol Lett. 2016;11:1885-1888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
13.  Murakami N, Mulvaney P, Danesh M, Abudayyeh A, Diab A, Abdel-Wahab N, Abdelrahim M, Khairallah P, Shirazian S, Kukla A, Owoyemi IO, Alhamad T, Husami S, Menon M, Santeusanio A, Blosser CD, Zuniga SC, Soler MJ, Moreso F, Mithani Z, Ortiz-Melo D, Jaimes EA, Gutgarts V, Lum E, Danovitch GM, Cardarelli F, Drews RE, Bassil C, Swank JL, Westphal S, Mannon RB, Shirai K, Kitchlu A, Ong S, Machado SM, Mothi SS, Ott PA, Rahma O, Hodi FS, Sise ME, Gupta S, Leaf DE, Devoe CE, Wanchoo R, Nair VV, Schmults CD, Hanna GJ, Sprangers B, Riella LV, Jhaveri KD; Immune Checkpoint Inhibitors in Solid Organ Transplant Consortium. A multi-center study on safety and efficacy of immune checkpoint inhibitors in cancer patients with kidney transplant. Kidney Int. 2021;100:196-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 80]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
14.  De Bruyn P, Van Gestel D, Ost P, Kruse V, Brochez L, Van Vlierberghe H, Devresse A, Del Marmol V, Le Moine A, Aspeslagh S. Immune checkpoint blockade for organ transplant patients with advanced cancer: how far can we go? Curr Opin Oncol. 2019;31:54-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 37]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
15.  Schvartsman G, Perez K, Sood G, Katkhuda R, Tawbi H. Immune Checkpoint Inhibitor Therapy in a Liver Transplant Recipient With Melanoma. Ann Intern Med. 2017;167:361-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 31]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
16.  Morales RE, Shoushtari AN, Walsh MM, Grewal P, Lipson EJ, Carvajal RD. Safety and efficacy of ipilimumab to treat advanced melanoma in the setting of liver transplantation. J Immunother Cancer. 2015;3:22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 83]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
17.  Finn RS, Ryoo BY, Merle P, Kudo M, Bouattour M, Lim HY, Breder V, Edeline J, Chao Y, Ogasawara S, Yau T, Garrido M, Chan SL, Knox J, Daniele B, Ebbinghaus SW, Chen E, Siegel AB, Zhu AX, Cheng AL; KEYNOTE-240 investigators. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 682]  [Cited by in F6Publishing: 1082]  [Article Influence: 216.4]  [Reference Citation Analysis (0)]
18.  Yau T, Park JW, Finn RS, Cheng AL, Mathurin P, Edeline J, Kudo M, Han KH, Harding JJ, Merle P, Rosmorduc O, Wyrwicz L, Schott E, Choo SP, Kelley RK, Begic D, Chen G, Neely J, Anderson J, Sangro B. CheckMate 459: A randomized, multi-center phase III study of nivolumab (NIVO) vs sorafenib (SOR) as first-line (1L) treatment in patients (pts) with advanced hepatocellular carcinoma (aHCC). Ann Oncol. 2019;30:874.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Hafezi M, Lin M, Chia A, Chua A, Ho ZZ, Fam R, Tan D, Aw J, Pavesi A, Krishnamoorthy TL, Chow WC, Chen W, Zhang Q, Wai LE, Koh S, Tan AT, Bertoletti A. Immunosuppressive Drug-Resistant Armored T-Cell Receptor T Cells for Immune Therapy of HCC in Liver Transplant Patients. Hepatology. 2021;74:200-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 26]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
20.  Qasim W, Brunetto M, Gehring AJ, Xue SA, Schurich A, Khakpoor A, Zhan H, Ciccorossi P, Gilmour K, Cavallone D, Moriconi F, Farzhenah F, Mazzoni A, Chan L, Morris E, Thrasher A, Maini MK, Bonino F, Stauss H, Bertoletti A. Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. J Hepatol. 2015;62:486-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 128]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
21.  Tan AT, Yang N, Lee Krishnamoorthy T, Oei V, Chua A, Zhao X, Tan HS, Chia A, Le Bert N, Low D, Tan HK, Kumar R, Irani FG, Ho ZZ, Zhang Q, Guccione E, Wai LE, Koh S, Hwang W, Chow WC, Bertoletti A. Use of Expression Profiles of HBV-DNA Integrated Into Genomes of Hepatocellular Carcinoma Cells to Select T Cells for Immunotherapy. Gastroenterology. 2019;156:1862-1876.e9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 70]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
22.  Xie S, Wu Z, Zhou L, Liang Y, Wang X, Niu L, Xu K, Chen J, Zhang M. Iodine-125 seed implantation and allogenic natural killer cell immunotherapy for hepatocellular carcinoma after liver transplantation: a case report. Onco Targets Ther. 2018;11:7345-7352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
23.  Kumar V, Shinagare AB, Rennke HG, Ghai S, Lorch JH, Ott PA, Rahma OE. The Safety and Efficacy of Checkpoint Inhibitors in Transplant Recipients: A Case Series and Systematic Review of Literature. Oncologist. 2020;25:505-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 82]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
24.  Amjad W, Kotiah S, Gupta A, Morris M, Liu L, Thuluvath PJ. Successful Treatment of Disseminated Hepatocellular Carcinoma After Liver Transplantation With Nivolumab. J Clin Exp Hepatol. 2020;10:185-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
25.  Varkaris A, Lewis DW, Nugent FW. Preserved Liver Transplant After PD-1 Pathway Inhibitor for Hepatocellular Carcinoma. Am J Gastroenterol. 2017;112:1895-1896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 46]  [Article Influence: 6.6]  [Reference Citation Analysis (1)]
26.  Munker S, De Toni EN. Use of checkpoint inhibitors in liver transplant recipients. United European Gastroenterol J. 2018;6:970-973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 64]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
27.  De Toni EN, Gerbes AL. Tapering of Immunosuppression and Sustained Treatment With Nivolumab in a Liver Transplant Recipient. Gastroenterology. 2017;152:1631-1633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
28.  Halloran PF. Immunosuppressive drugs for kidney transplantation. N Engl J Med. 2004;351:2715-2729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1087]  [Cited by in F6Publishing: 980]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
29.  Ho CM, Chen HL, Hu RH, Lee PH. Harnessing immunotherapy for liver recipients with hepatocellular carcinoma: a review from a transplant oncology perspective. Ther Adv Med Oncol. 2019;11:1758835919843463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
30.  Diehl R, Ferrara F, Müller C, Dreyer AY, McLeod DD, Fricke S, Boltze J. Immunosuppression for in vivo research: state-of-the-art protocols and experimental approaches. Cell Mol Immunol. 2017;14:146-179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 91]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
31.  Geissler EK, Schlitt HJ. Immunosuppression for liver transplantation. Gut. 2009;58:452-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 103]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
32.  Rammohan A, Reddy MS, Farouk M, Vargese J, Rela M. Pembrolizumab for metastatic hepatocellular carcinoma following live donor liver transplantation: The silver bullet? Hepatology. 2018;67:1166-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 50]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
33.  Klawitter J, Nashan B, Christians U. Everolimus and sirolimus in transplantation-related but different. Expert Opin Drug Saf. 2015;14:1055-1070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 96]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
34.  Wong M. Mammalian target of rapamycin (mTOR) pathways in neurological diseases. Biomed J. 2013;36:40-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 117]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
35.  Schnitzbauer AA, Filmann N, Adam R, Bachellier P, Bechstein WO, Becker T, Bhoori S, Bilbao I, Brockmann J, Burra P, Chazoullières O, Cillo U, Colledan M, Duvoux C, Ganten TM, Gugenheim J, Heise M, van Hoek B, Jamieson N, de Jong KP, Klein CG, Klempnauer J, Kneteman N, Lerut J, Mäkisalo H, Mazzaferro V, Mirza DF, Nadalin S, Neuhaus P, Pageaux GP, Pinna AD, Pirenne J, Pratschke J, Powel J, Rentsch M, Rizell M, Rossi G, Rostaing L, Roy A, Scholz T, Settmacher U, Soliman T, Strasser S, Söderdahl G, Troisi RI, Turrión VS, Schlitt HJ, Geissler EK. mTOR Inhibition Is Most Beneficial After Liver Transplantation for Hepatocellular Carcinoma in Patients With Active Tumors. Ann Surg. 2020;272:855-862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 74]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
36.  Esfahani K, Al-Aubodah TA, Thebault P, Lapointe R, Hudson M, Johnson NA, Baran D, Bhulaiga N, Takano T, Cailhier JF, Piccirillo CA, Miller WH. Targeting the mTOR pathway uncouples the efficacy and toxicity of PD-1 blockade in renal transplantation. Nat Commun. 2019;10:4712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 68]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
37.  Biondani P, De Martin E, Samuel D. Safety of an anti-PD-1 immune checkpoint inhibitor in a liver transplant recipient. Ann Oncol. 2018;29:286-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 36]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
38.  Abdel-Wahab N, Safa H, Abudayyeh A, Johnson DH, Trinh VA, Zobniw CM, Lin H, Wong MK, Abdelrahim M, Gaber AO, Suarez-Almazor ME, Diab A. Checkpoint inhibitor therapy for cancer in solid organ transplantation recipients: an institutional experience and a systematic review of the literature. J Immunother Cancer. 2019;7:106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 172]  [Article Influence: 34.4]  [Reference Citation Analysis (0)]
39.  Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/ B7 family. Immunol Rev. 2011;241:180-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 285]  [Cited by in F6Publishing: 301]  [Article Influence: 23.2]  [Reference Citation Analysis (0)]
40.  Tanaka K, Albin MJ, Yuan X, Yamaura K, Habicht A, Murayama T, Grimm M, Waaga AM, Ueno T, Padera RF, Yagita H, Azuma M, Shin T, Blazar BR, Rothstein DM, Sayegh MH, Najafian N. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. J Immunol. 2007;179:5204-5210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 153]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
41.  Yu S, Su C, Luo X. Impact of infection on transplantation tolerance. Immunol Rev. 2019;292:243-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
42.  Fisher J, Zeitouni N, Fan W, Samie FH. Immune checkpoint inhibitor therapy in solid organ transplant recipients: A patient-centered systematic review. J Am Acad Dermatol. 2020;82:1490-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 68]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
43.  Aguirre LE, Guzman ME, Lopes G, Hurley J. Immune Checkpoint Inhibitors and the Risk of Allograft Rejection: A Comprehensive Analysis on an Emerging Issue. Oncologist. 2019;24:394-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
44.  Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev. 2008;224:166-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 626]  [Cited by in F6Publishing: 691]  [Article Influence: 43.2]  [Reference Citation Analysis (0)]
45.  Yoo SH, Keam B, Kim M, Kim SH, Kim YJ, Kim TM, Kim DW, Lee JS, Heo DS. Low-dose nivolumab can be effective in non-small cell lung cancer: alternative option for financial toxicity. ESMO Open. 2018;3:e000332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 49]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
46.  Agrawal S, Feng Y, Roy A, Kollia G, Lestini B. Nivolumab dose selection: challenges, opportunities, and lessons learned for cancer immunotherapy. J Immunother Cancer. 2016;4:72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 134]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
47.  El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, Kim TY, Choo SP, Trojan J, Welling TH Rd, Meyer T, Kang YK, Yeo W, Chopra A, Anderson J, Dela Cruz C, Lang L, Neely J, Tang H, Dastani HB, Melero I. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2536]  [Cited by in F6Publishing: 2865]  [Article Influence: 409.3]  [Reference Citation Analysis (1)]
48.  Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, Gilson MM, Wang C, Selby M, Taube JM, Anders R, Chen L, Korman AJ, Pardoll DM, Lowy I, Topalian SL. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2135]  [Cited by in F6Publishing: 2271]  [Article Influence: 162.2]  [Reference Citation Analysis (0)]
49.  Pandey A, Cohen DJ. Ipilumumab for hepatocellular cancer in a liver transplant recipient, with durable response, tolerance and without allograft rejection. Immunotherapy. 2020;12:287-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
50.  Sun B, Yang D, Dai H, Liu X, Jia R, Cui X, Li W, Cai C, Xu J, Zhao X. Eradication of Hepatocellular Carcinoma by NKG2D-Based CAR-T Cells. Cancer Immunol Res. 2019;7:1813-1823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
51.  Shi D, Shi Y, Kaseb AO, Qi X, Zhang Y, Chi J, Lu Q, Gao H, Jiang H, Wang H, Yuan D, Ma H, Li Z, Zhai B. Chimeric Antigen Receptor-Glypican-3 T-Cell Therapy for Advanced Hepatocellular Carcinoma: Results of Phase I Trials. Clin Cancer Res. 2020;26:3979-3989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 161]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
52.  Loh K, Khan K, Kroemer A. CAR-NK Therapy: Translating a Novel Immunotherapy Treatment to Solid Organ Transplantation. Transplantation. 2020;104:1523-1524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
53.  Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, Chen C, Olive O, Carter TA, Li S, Lieb DJ, Eisenhaure T, Gjini E, Stevens J, Lane WJ, Javeri I, Nellaiappan K, Salazar AM, Daley H, Seaman M, Buchbinder EI, Yoon CH, Harden M, Lennon N, Gabriel S, Rodig SJ, Barouch DH, Aster JC, Getz G, Wucherpfennig K, Neuberg D, Ritz J, Lander ES, Fritsch EF, Hacohen N, Wu CJ. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1520]  [Cited by in F6Publishing: 1741]  [Article Influence: 248.7]  [Reference Citation Analysis (0)]
54.  Keskin DB, Anandappa AJ, Sun J, Tirosh I, Mathewson ND, Li S, Oliveira G, Giobbie-Hurder A, Felt K, Gjini E, Shukla SA, Hu Z, Li L, Le PM, Allesøe RL, Richman AR, Kowalczyk MS, Abdelrahman S, Geduldig JE, Charbonneau S, Pelton K, Iorgulescu JB, Elagina L, Zhang W, Olive O, McCluskey C, Olsen LR, Stevens J, Lane WJ, Salazar AM, Daley H, Wen PY, Chiocca EA, Harden M, Lennon NJ, Gabriel S, Getz G, Lander ES, Regev A, Ritz J, Neuberg D, Rodig SJ, Ligon KL, Suvà ML, Wucherpfennig KW, Hacohen N, Fritsch EF, Livak KJ, Ott PA, Wu CJ, Reardon DA. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565:234-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 824]  [Cited by in F6Publishing: 803]  [Article Influence: 160.6]  [Reference Citation Analysis (0)]
55.  Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, Budde LE, Costa L, Davies M, Dunnington D, Ernstoff MS, Frigault M, Hoffner B, Hoimes CJ, Lacouture M, Locke F, Lunning M, Mohindra NA, Naidoo J, Olszanski AJ, Oluwole O, Patel SP, Reddy S, Ryder M, Santomasso B, Shofer S, Sosman JA, Wahidi M, Wang Y, Johnson-Chilla A, Scavone JL. Management of Immunotherapy-Related Toxicities, Version 1.2019. J Natl Compr Canc Netw. 2019;17:255-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 288]  [Article Influence: 57.6]  [Reference Citation Analysis (0)]
56.  Remash D, Prince DS, McKenzie C, Strasser SI, Kao S, Liu K. Immune checkpoint inhibitor-related hepatotoxicity: A review. World J Gastroenterol. 2021;27:5376-5391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (1)]
57.  Demetris AJ, Ruppert K, Dvorchik I, Jain A, Minervini M, Nalesnik MA, Randhawa P, Wu T, Zeevi A, Abu-Elmagd K, Eghtesad B, Fontes P, Cacciarelli T, Marsh W, Geller D, Fung JJ. Real-time monitoring of acute liver-allograft rejection using the Banff schema. Transplantation. 2002;74:1290-1296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 80]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  Gassmann D, Weiler S, Mertens JC, Reiner CS, Vrugt B, Nägeli M, Mangana J, Müllhaupt B, Jenni F, Misselwitz B. Liver Allograft Failure After Nivolumab Treatment-A Case Report With Systematic Literature Research. Transplant Direct. 2018;4:e376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 84]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
59.  Friend BD, Venick RS, McDiarmid SV, Zhou X, Naini B, Wang H, Farmer DG, Busuttil RW, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 89]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
60.  DeLeon TT, Salomao MA, Aqel BA, Sonbol MB, Yokoda RT, Ali AH, Moss AA, Mathur AK, Chascsa DM, Rakela J, Bryce AH, Borad MJ. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 94]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
61.  Lefaucheur C, Loupy A, Vernerey D, Duong-Van-Huyen JP, Suberbielle C, Anglicheau D, Vérine J, Beuscart T, Nochy D, Bruneval P, Charron D, Delahousse M, Empana JP, Hill GS, Glotz D, Legendre C, Jouven X. Antibody-mediated vascular rejection of kidney allografts: a population-based study. Lancet. 2013;381:313-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 253]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
62.  Lipson EJ, Bagnasco SM, Moore J Jr, Jang S, Patel MJ, Zachary AA, Pardoll DM, Taube JM, Drake CG. Tumor Regression and Allograft Rejection after Administration of Anti-PD-1. N Engl J Med. 2016;374:896-898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 214]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
63.  Adams DH, Neuberger JM. Treatment of acute rejection. Semin Liver Dis. 1992;12:80-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
64.  Kamar N, Lavayssière L, Muscari F, Selves J, Guilbeau-Frugier C, Cardeau I, Esposito L, Cointault O, Nogier MB, Peron JM, Otal P, Fort M, Rostaing L. Early plasmapheresis and rituximab for acute humoral rejection after ABO-compatible liver transplantation. World J Gastroenterol. 2009;15:3426-3430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 38]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
65.  Schmitt TM, Phillips M, Sawyer RG, Northup P, Hagspiel KD, Pruett TL, Bonatti HJ. Anti-thymocyte globulin for the treatment of acute cellular rejection following liver transplantation. Dig Dis Sci. 2010;55:3224-3234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
66.  Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 544]  [Cited by in F6Publishing: 582]  [Article Influence: 72.8]  [Reference Citation Analysis (0)]
67.  Dromi SA, Walsh MP, Herby S, Traughber B, Xie J, Sharma KV, Sekhar KP, Luk A, Liewehr DJ, Dreher MR, Fry TJ, Wood BJ. Radiofrequency ablation induces antigen-presenting cell infiltration and amplification of weak tumor-induced immunity. Radiology. 2009;251:58-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 160]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
68.  Benson AB, D'Angelica MI, Abbott DE, Anaya DA, Anders R, Are C, Bachini M, Borad M, Brown D, Burgoyne A, Chahal P, Chang DT, Cloyd J, Covey AM, Glazer ES, Goyal L, Hawkins WG, Iyer R, Jacob R, Kelley RK, Kim R, Levine M, Palta M, Park JO, Raman S, Reddy S, Sahai V, Schefter T, Singh G, Stein S, Vauthey JN, Venook AP, Yopp A, McMillian NR, Hochstetler C, Darlow SD. Hepatobiliary Cancers, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2021;19:541-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 386]  [Article Influence: 128.7]  [Reference Citation Analysis (0)]
69.  National Comprehensive Cancer Network  (NCCN) Clinical Practice Guidelines in Oncology. Hepatobiliary Cancer, Version 3. 2021. [cited 20 June 2021]. In: National Comprehensive Cancer Network [Internet]. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1438.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Finn RS, Ikeda M, Zhu AX, Sung MW, Baron AD, Kudo M, Okusaka T, Kobayashi M, Kumada H, Kaneko S, Pracht M, Mamontov K, Meyer T, Kubota T, Dutcus CE, Saito K, Siegel AB, Dubrovsky L, Mody K, Llovet JM. Phase Ib Study of Lenvatinib Plus Pembrolizumab in Patients With Unresectable Hepatocellular Carcinoma. J Clin Oncol. 2020;38:2960-2970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 661]  [Cited by in F6Publishing: 654]  [Article Influence: 163.5]  [Reference Citation Analysis (0)]
71.  Cheng AL, Hsu C, Chan SL, Choo SP, Kudo M. Challenges of combination therapy with immune checkpoint inhibitors for hepatocellular carcinoma. J Hepatol. 2020;72:307-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 277]  [Article Influence: 69.3]  [Reference Citation Analysis (1)]
72.  Farsaci B, Donahue RN, Coplin MA, Grenga I, Lepone LM, Molinolo AA, Hodge JW. Immune consequences of decreasing tumor vasculature with antiangiogenic tyrosine kinase inhibitors in combination with therapeutic vaccines. Cancer Immunol Res. 2014;2:1090-1102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 54]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
73.  Chen ML, Yan BS, Lu WC, Chen MH, Yu SL, Yang PC, Cheng AL. Sorafenib relieves cell-intrinsic and cell-extrinsic inhibitions of effector T cells in tumor microenvironment to augment antitumor immunity. Int J Cancer. 2014;134:319-331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 94]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
74.  Sunay MM, Foote JB, Leatherman JM, Edwards JP, Armstrong TD, Nirschl CJ, Hicks J, Emens LA. Sorafenib combined with HER-2 targeted vaccination can promote effective T cell immunity in vivo. Int Immunopharmacol. 2017;46:112-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
75.  Ren Z, Li A, Jiang J, Zhou L, Yu Z, Lu H, Xie H, Chen X, Shao L, Zhang R, Xu S, Zhang H, Cui G, Sun R, Wen H, Lerut JP, Kan Q, Li L, Zheng S. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut. 2019;68:1014-1023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in F6Publishing: 387]  [Article Influence: 77.4]  [Reference Citation Analysis (0)]
76.  Kato K, Nagao M, Miyamoto K, Oka K, Takahashi M, Yamamoto M, Matsumura Y, Kaido T, Uemoto S, Ichiyama S. Longitudinal Analysis of the Intestinal Microbiota in Liver Transplantation. Transplant Direct. 2017;3:e144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 46]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
77.  Sun LY, Yang YS, Qu W, Zhu ZJ, Wei L, Ye ZS, Zhang JR, Sun XY, Zeng ZG. Gut microbiota of liver transplantation recipients. Sci Rep. 2017;7:3762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 38]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
78.  Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, Cogdill AP, Zhao L, Hudgens CW, Hutchinson DS, Manzo T, Petaccia de Macedo M, Cotechini T, Kumar T, Chen WS, Reddy SM, Szczepaniak Sloane R, Galloway-Pena J, Jiang H, Chen PL, Shpall EJ, Rezvani K, Alousi AM, Chemaly RF, Shelburne S, Vence LM, Okhuysen PC, Jensen VB, Swennes AG, McAllister F, Marcelo Riquelme Sanchez E, Zhang Y, Le Chatelier E, Zitvogel L, Pons N, Austin-Breneman JL, Haydu LE, Burton EM, Gardner JM, Sirmans E, Hu J, Lazar AJ, Tsujikawa T, Diab A, Tawbi H, Glitza IC, Hwu WJ, Patel SP, Woodman SE, Amaria RN, Davies MA, Gershenwald JE, Hwu P, Lee JE, Zhang J, Coussens LM, Cooper ZA, Futreal PA, Daniel CR, Ajami NJ, Petrosino JF, Tetzlaff MT, Sharma P, Allison JP, Jenq RR, Wargo JA. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2846]  [Cited by in F6Publishing: 2679]  [Article Influence: 446.5]  [Reference Citation Analysis (0)]
79.  Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, Fidelle M, Flament C, Poirier-Colame V, Opolon P, Klein C, Iribarren K, Mondragón L, Jacquelot N, Qu B, Ferrere G, Clémenson C, Mezquita L, Masip JR, Naltet C, Brosseau S, Kaderbhai C, Richard C, Rizvi H, Levenez F, Galleron N, Quinquis B, Pons N, Ryffel B, Minard-Colin V, Gonin P, Soria JC, Deutsch E, Loriot Y, Ghiringhelli F, Zalcman G, Goldwasser F, Escudier B, Hellmann MD, Eggermont A, Raoult D, Albiges L, Kroemer G, Zitvogel L. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2493]  [Cited by in F6Publishing: 3120]  [Article Influence: 445.7]  [Reference Citation Analysis (0)]
80.  Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, Rotin D, Anafi L, Avivi C, Melnichenko J, Steinberg-Silman Y, Mamtani R, Harati H, Asher N, Shapira-Frommer R, Brosh-Nissimov T, Eshet Y, Ben-Simon S, Ziv O, Khan MAW, Amit M, Ajami NJ, Barshack I, Schachter J, Wargo JA, Koren O, Markel G, Boursi B. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:602-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 680]  [Article Influence: 170.0]  [Reference Citation Analysis (0)]
81.  Lu S, Stein JE, Rimm DL, Wang DW, Bell JM, Johnson DB, Sosman JA, Schalper KA, Anders RA, Wang H, Hoyt C, Pardoll DM, Danilova L, Taube JM. Comparison of Biomarker Modalities for Predicting Response to PD-1/PD-L1 Checkpoint Blockade: A Systematic Review and Meta-analysis. JAMA Oncol. 2019;5:1195-1204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 366]  [Cited by in F6Publishing: 377]  [Article Influence: 75.4]  [Reference Citation Analysis (0)]
82.  Yau T, Kang YK, Kim TY, El-Khoueiry AB, Santoro A, Sangro B, Melero I, Kudo M, Hou MM, Matilla A, Tovoli F, Knox JJ, Ruth He A, El-Rayes BF, Acosta-Rivera M, Lim HY, Neely J, Shen Y, Wisniewski T, Anderson J, Hsu C. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 505]  [Cited by in F6Publishing: 671]  [Article Influence: 167.8]  [Reference Citation Analysis (0)]
83.  Xu X, Tan Y, Qian Y, Xue W, Wang Y, Du J, Jin L, Ding W. Clinicopathologic and prognostic significance of tumor-infiltrating CD8+ T cells in patients with hepatocellular carcinoma: A meta-analysis. Medicine (Baltimore). 2019;98:e13923.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
84.  Duffy AG, Ulahannan SV, Makorova-Rusher O, Rahma O, Wedemeyer H, Pratt D, Davis JL, Hughes MS, Heller T, ElGindi M, Uppala A, Korangy F, Kleiner DE, Figg WD, Venzon D, Steinberg SM, Venkatesan AM, Krishnasamy V, Abi-Jaoudeh N, Levy E, Wood BJ, Greten TF. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J Hepatol. 2017;66:545-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 454]  [Cited by in F6Publishing: 557]  [Article Influence: 79.6]  [Reference Citation Analysis (0)]
85.  Tumeh PC, Harview CL, Yearley JH, Shintaku IP, Taylor EJ, Robert L, Chmielowski B, Spasic M, Henry G, Ciobanu V, West AN, Carmona M, Kivork C, Seja E, Cherry G, Gutierrez AJ, Grogan TR, Mateus C, Tomasic G, Glaspy JA, Emerson RO, Robins H, Pierce RH, Elashoff DA, Robert C, Ribas A. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4180]  [Cited by in F6Publishing: 4769]  [Article Influence: 529.9]  [Reference Citation Analysis (0)]
86.  Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, Hodi FS, Martín-Algarra S, Mandal R, Sharfman WH, Bhatia S, Hwu WJ, Gajewski TF, Slingluff CL Jr, Chowell D, Kendall SM, Chang H, Shah R, Kuo F, Morris LGT, Sidhom JW, Schneck JP, Horak CE, Weinhold N, Chan TA. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell. 2017;171:934-949.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 978]  [Cited by in F6Publishing: 1278]  [Article Influence: 182.6]  [Reference Citation Analysis (0)]
87.  Patel P, Schutzer SE, Pyrsopoulos N. Immunobiology of hepatocarcinogenesis: Ways to go or almost there? World J Gastrointest Pathophysiol. 2016;7:242-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
88.  Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, Majem M, Fidler MJ, de Castro G Jr, Garrido M, Lubiniecki GM, Shentu Y, Im E, Dolled-Filhart M, Garon EB. Pembrolizumab vs docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387:1540-1550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4116]  [Cited by in F6Publishing: 4661]  [Article Influence: 582.6]  [Reference Citation Analysis (0)]
89.  Callahan MK, Postow MA, Wolchok JD. Immunomodulatory therapy for melanoma: ipilimumab and beyond. Clin Dermatol. 2013;31:191-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
90.  Zhuang L, Mou HB, Yu LF, Zhu HK, Yang Z, Liao Q, Zheng SS. Immune checkpoint inhibitor for hepatocellular carcinoma recurrence after liver transplantation. Hepatobiliary Pancreat Dis Int. 2020;19:91-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
91.  Al Jarroudi O, Ulusakarya A, Almohamad W, Afqir S, Morere JF. Anti-Programmed Cell Death Protein 1 (PD-1) Immunotherapy for Metastatic Hepatocellular Carcinoma After Liver Transplantation: A Report of Three Cases. Cureus. 2020;12:e11150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
92.  Wang GY, Tang H, Zhang YC, Li H, Yi SH, Jiang N, Wang GS, Zhang J, Zhang Q, Yang Y, Chen GH. Programmed death receptor (PD) -1 monoclonal antibody-induced acute immune hepatitis in the treatment of recurrent hepatocellular carcinoma after liver transplantation: a case report. Organ Transplant. 2016;1:44-47.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Qiu J, Tang W, Du C. Immune Checkpoint Inhibitors in Patients with Recurrent Hepatocellular Carcinoma after Liver Transplantation: A Case Report and Literature Review. Curr Cancer Drug Targets. 2020;20:720-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]