v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Hepatology > Article Full Text

  • OPEN ACCESS

Hepatocellular Carcinoma: Downstaging to Liver Transplantation as Curative Therapy

  • Leana Frankul  and
  • Catherine Frenette* 
 Author information
Journal of Clinical and Translational Hepatology   2021;9(2):220-226

doi: 10.14218/JCTH.2020.00037

Abstract

Hepatocellular carcinoma (HCC) ranks among the leading cancer-related causes of morbidity and mortality worldwide. Downstaging of HCC has prevailed as a key method to curative therapy for patients who present with unresectable HCC outside of the listing criteria for liver transplantation (LT). Even though LT paves the way to lifesaving curative therapy for HCC, perpetually severe organ shortage limits its broader application. Debate over the optimal protocol and assessment of response to downstaging treatment has fueled immense research activity and is pushing the boundaries of LT candidate selection criteria. The implicit obligation of refining downstaging protocol is to ensure the maximization of the transplant survival benefit by taking into account the waitlist life expectancy. In the following review, we critically discuss strategies to best optimize downstaging HCC to LT on the basis of existing literature.

Keywords

Downstaging, Milan criteria, Hepatocellular carcinoma, Liver transplantation

Introduction

Hepatocellular carcinoma (HCC) is the most prevalent primary liver malignancy. It is the sixth most common neoplasm and fourth cause of cancer-related mortality globally.1,2 As the incidence of HCC is projected to increase in the USA as nonalcoholic fatty liver disease continues to increase exponentially and alcohol and hepatitis C remain public health issues, HCC has emerged as a leading indication for liver transplantation (LT).3–5

LT offers a successful therapy for early-stage HCC patients because it simultaneously removes the lesion(s) and the preneoplastic liver.6 Early records of post-LT outcome delineated high recurrence rates and were plagued with dismal patient survival.7,8 Apart from tumor measurements, factors influencing recurrence include vascular invasion, histologic differentiation, previous response to local-regional therapy (LRT) and serum marker levels.9–12

The primary aims of establishing criteria for LT are to select candidates with good post-LT prognoses and to exclude patients whose disease conditions are suitable for other therapies, such as resection or systemic therapy. The Milan criteria (MC) (a single nodule ≤5 cm, 2–3 nodules ≤3 cm), proposed in 1996, emerged as an international benchmark to select patients with HCC for LT. According to MC, post-LT 5-year survival in HCC is >70% with a recurrence rate <10–15%.13–15 The American Association for the Study of Liver Disease (commonly known as AASLD) and Guidelines of the European Association for the Study of the Liver (commonly known as EASL) recommend LT for HCC patients within MC but unsuitable for resection.16,17

However, debate in the past two decades has revolved around the dichotomous nature of MC. The stringent MC precludes access to LT for patients with larger or more numerous tumors who potentially have acceptable post-LT outcomes but who otherwise are not candidates for curative therapy. A plethora of studies have evaluated the liberalization from conventional criteria for HCC LT.18–24 An alternative form of expansion relates to LT of candidates whose tumor burden exceeds MC without utilizing pre-LT treatment, while another form is linked to using treatment to successfully “downstage” tumor burden to within standard LT listing criteria based on radiographic assessment and markers of tumor biology. The current article reviews the framework for the downstaging of HCC and sheds light on recent updates in the field of prognosticators of post-LT outcomes.

Expanded selection criteria

Several expanded criteria for HCC beyond MC have been proposed (Table 1).13–15,19–24 It is important to preface that most of the earlier studies predominantly relied on tumor morphological characteristics, which undermined their power in establishing ideal cutoffs. Additionally, prospective study design constructs a stronger evidential foundation for expanded criteria than does retrospective study proposals, by eliminating confounding variables and carefully selecting participants.25,26

Table 1

Details of different criteria for LT in HCC

Selection systemAssessmentCriteriaYears of follow-upSurvival, %Recurrence rate, %
MC1315RadiologyTumor size of ≤5 cm; up to three separate lesions, none larger than 3 cm; no evidence of gross vascular invasion; and no regional nodal or distant metastases4>70 (OS)<10–15
UCSF criteria19RadiologySingle tumor ≤6.5 cm or two to three lesions, none exceeding 4.5 cm with total tumor diameter ≤8 cm580.9 (RFS)9.1
Total tumor volume and AFP criteria21RadiologyTotal tumor volume ≤115 cm3 and AFP ≤400 ng/mL, without macrovascular invasion474.6 (OS)9.4
Up-to-seven criteria20PathologySize of largest HCC plus number of HCCs ≤7571.2 (OS)9.1
ETC24RadiologyAny size or number of tumors, provided no extrahepatic spread, vascular invasion, or poor differentiation on pre-LT biopsy568 (OS)25.6
Hangzhou criteria22PathologyTotal tumor diameter ≤8 cm or a total tumor diameter >8 cm, with a histopathologic grade I or II and a preoperative AFP ≤400 ng/mL570.7 (OS)N/A
Kyoto criteria23RadiologyTumor number ≤10; all ≤5 cm; and serum DCP ≤400 mAU /mL565 (OS)30

In 2001, Yao et al.18 retrospectively analyzed LT recipients and propounded a modestly expanded criteria for orthotopic liver transplantation (referred to herein as OLT) on the grounds of explant histological characteristics. The exploratory study set forth the University of California in San Francisco (UCSF) criteria: (1) single lesion ≤6.5 cm; or (2) ≤3 lesions, with the largest ≤4.5 cm and total sum of diameters ≤8 cm. In a follow-up study, Yao et al.19 prospectively validated the UCSF criteria for OLT based on pretransplant imaging and outlined post-OLT tumor recurrence and survival. The 5-year patient survival without recurrence was 81% and the recurrence-free probability exceeded 90% for patients meeting the UCSF criteria, which were similar to the patients fulfilling the MC.

Mazzaferro et al.20 examined the feasibility of “up-to-seven criteria” (the sum of the size of the largest nodule and the number of nodules ≤7 without microvascular invasion) derived from explant pathology collected from 36 centers worldwide. Notably, the 71.2% 5-year OS rate achieved among patients beyond MC but within the “up-to-seven” criteria was associated with the absence of microvascular invasion, a variable difficult to ascertain pre-LT. It is noteworthy that upper tumor size and number limits beyond MC may increase the likelihood of microvascular invasion.27

In a prospective validation attempt to extend MC, Toso et al.21 presented data in which LT candidate selection depended on a composite of the total tumor volume (≤115 cm3) and alpha-fetoprotein (AFP) ≤400 ng/mL without macrovascular invasion or extrahepatic disease. Even though post-LT survival and recurrence were comparable to patients meeting MC, the waitlist drop-out rates posed a disadvantage. In China, the Hangzhou criteria also accounted for AFP levels in their protocol for selecting HCC patients for LT. Specifically, the Hangzhou criteria integrated total tumor diameter ≤8 cm or a total tumor diameter >8 cm, with a histopathologic grade I or II and preoperative AFP ≤400 ng/mL.22

A research group from Kyoto University proposed the Kyoto criteria that involved HCC tumor number ≤10, each tumor diameter no larger than 5 cm, and serum des-γ-carboxy prothrombin (referred to herein as DCP) ≤400 mAU /mL. The group’s recent intention to treat analysis resulted in a 5-year OS rate and recurrence rate of 82% and 7%, respectively.23

Researchers at the University of Toronto endeavored to validate their extended Toronto criteria (commonly known as ETC), which relied on poor tumor differentiation, elevated AFP and cancer-related symptoms to select HCC candidates for LT, rather than the conventional measurements of tumor size and number at presentation. Although the 5-year OS of 68% for patients transplanted according to ETC was not statistically inferior to patients within MC amongst the prospective cohort of patients followed, tumor recurrence post-LT was higher for patients who exceeded MC but satisfied the ETC.24

Nonetheless, MC remains the gold standard for HCC patient selection and prognostic evaluation in LT.28 The adoption of extended selection criteria generates the dilemma of a sharp rise in HCC patients on the LT waitlist with unknown regional repercussions on non-HCC patients waiting for LT, while persistent shortages of donor organs highlight the fundamental challenge of maintaining equity in liver transplant allocation.

Dynamism of serum markers

The multifactorial nature of HCC necessitates the integration of prognostic markers to assess tumor biological features and vascular invasion during the transplant evaluation process. No longer a contentious tool in candidate selection, AFP is widely used to distinguish the subset of LT candidates with a reasonable life expectancy after LT.29–32 Many liver transplant centers globally incorporate AFP into their listing criteria, with differences in cutoffs. Therefore, the optimal serum AFP level cutoff as an exclusion criterion for LT in pre-transplant HCC patients has garnered conspicuous research focus. In a detailed analysis of national United Network for Organ Sharing (UNOS) data, the subset of patients outside the MC with low serum AFP levels (0–15 ng/mL) displayed improved post-LT survival.30 The high end of AFP level cutoff ranges from 400 ng/mL to 1,000 ng/mL.33–35 Mounting evidence reveals that AFP >1,000 ng/mL manifested in HCC patients either within or outside MC portends reduced post-LT survival and considerable risk for HCC recurrence.34,36,37

There is a paucity of data on the predictive value of other serum markers for post-transplant mortality and HCC recurrence. In the absence of a universal AFP cutoff point, some members of the liver transplant community have investigated DCP, lens culinaris agglutinin-reactive alpha-fetoprotein (AFP-L3) and/or the ratio of AFP-L3 to total AFP (AFP-L3%) as adjuncts within patient selection algorithms.38–40 Moreover, the elevated neutrophil-lymphocyte ratio, an index of systemic inflammation, has been pursued as a maker of propensity to recurrence and unfavorable prognosis in parallel with AFP.41 External validation is needed prior to amending organ allocation strategies to embrace these promising serum markers.

LRT: Bridging and downstaging

LRT plays a pivotal role in the therapeutic management of HCC patients. Forms of LRT encompass a wide range of modalities that include transarterial chemoembolization (TACE), radiofrequency ablation (RFA), microwave ablation (commonly known as MWA), radioembolization, stereotactic body radiotherapy (commonly known as SBRT) and/or hepatic resection.42,43

LRT is frequently employed as a bridge to transplant in patients listed for LT within the Organ Procurement and Transplantation Network (commonly known as OPTN) T2 (Milan) criteria to prevent dropout from the waiting list by inducing tumor necrosis and deterring tumor progression.44,45 The rationale for bridging therapy lies in noncomparative studies reporting waitlist dropout rates as low as 8.7% at 6 months and between 22.9% at 12 months. By comparison, reported waitlist dropout rates are as high as 25% at 6 months and 38% at 12 months without the use of LRT.44,46–48 The possible beneficial effect of bridging therapy for HCC patients’ waitlist times of <6 months remains poorly characterized.28 Despite the liability for selection bias and lack of randomized control trials, European guidelines recommend LRT to reduce the risk of pre-LT drop-out in regions of anticipated wait times longer than 6 months.17 It is imperative to consider the risk of hepatic decompensation in advance of undergoing LRT. Furthermore, the variability in organ availability and hence vastly differing median waiting times across geographic regions culminate in a conditional recommendation for bridging therapy. Consequently, studies exploring LT waitlist dropout and post-LT outcomes founded on pre-transplantation treatment response radiologically evaluated by modified Response Evaluation Criteria in Solid Tumors (commonly known as mRECIST) are more logistically plausible to conduct than randomized controlled trials to elucidate the net effects of bridging LRT.49,50 Such future studies will also lend insight into how the development of new lesion(s) notwithstanding partial or complete response of the target lesion(s) affects outcomes. With the changes in UNOS model for end-stage liver disease score exception criteria now mandating a 6-month delay before exception points can be obtained, LRT has become standard of care in patients with HCC awaiting liver transplant. These changes inevitably cause a prolonged wait time that reinforces the usefulness of LRT. In a multivariate analysis of the UNOS database, Halazun et al.5 demonstrated that a waiting time of less than 4 to 6 months adversely impacts post-LT survival. Transplantation of patients with aggressive tumors in areas without a mandatory observational period can theoretically occur prematurely before tumor biologic behavior is assessed, thereby causing poor outcomes with aggressive recurrence. Accordingly, a minimal observation period aids in better candidate selection and possibly leads to lower risks of post-LT HCC recurrence.51,52

Tumor “downstaging” is a process that applies LRT to decrease tumor size and number in patients first deemed outside of the locally predefined criteria, commonly MC, for LT.53 First recommended in 1997, tumor downstaging provides a viable alternative approach to expanding MC limits to select a subgroup of patients whose LT candidacy would otherwise be disregarded.54–56 Sustained response to LRT can function as a measure of favorable tumor biology, whereas unresponsive and proliferative tumor burden after LRT yields worse post-LT outcomes.45,57–60 The latest AASLD guidelines suggest that patients beyond the MC (T3) should be considered for LT after successful downstaging to MC.16 Due to non-standardized downstaging protocol with precisely defined upper tumor limits across geographic regions, UNOS adopted the UCSF inclusion criteria for down-staging (single nodule ≤8 cm, 2–3 nodules each ≤5 cm, or 4–5 nodules each ≤3 cm with sum of the maximal tumor diameters ≤8 cm) as USA policy in 2017.58,61 The notion of placing restrictions to enter downstaging is predicated on concerns over fairness and appropriate prioritization in liver allocation for all indications.

The first analysis of the UNOS database of 3,276 patients within MC and 422 patients within UNOS downstaging criteria, who underwent LT from 2012 to 2015, confirmed the validity of UNOS downstaging criteria by showing similar 3-year post-LT survival between HCC patients always meeting MC and patients whose initial tumor burden met the UNOS downstaging criteria and were then downstaged to LT.62 Given the study’s dependence on pre-LT data submitted to UNOS by LT centers, reporting biases pertaining to radiographic response to LRT are plausible. For example, underestimation of tumor size, whether intentional or unintentional, can inflate the proportion of patients in the downstaging group with explant tumor burden beyond MC.63 Nevertheless, the findings that AFP ≥100 ng/mL at LT and short wait regions (median wait time of 2.6 months) or mid wait regions (median wait time of 6.5 months) were predictors for impaired post-LT survival in the downstaging groups support the need to incorporate AFP and expected wait times into tumor downstaging models.62

Efficacy of downstaging modalities

Currently, there are sparse data to draw conclusions on the optimal form of LRT for downstaging. Reported efficacies of common downstaging techniques defined as the successful anatomical reduction of tumor burden to within MC are highly variable.64–67 A systematic review by Parikh et al.65 revealed an overall downstaging success rate of 48%, with a post-LT HCC recurrence rate of 16%. The discrepancies in success of downstaging are attributed to various factors, such as initial tumor burden, choice of LRT utilized, LT program’s downstaging procedures, and lack of a standardized time interval to determine radiographic response to LRT. The type of LRT performed for each patient is contingent upon the location of tumor, underlying liver function, performance status of patient, as well as local expertise in each treatment modality. In this systematic review, there was no significant difference comparing TACE and transarterial radioembolization, but the highest success rates were in patients that underwent multimodal therapy. There was not a significant difference in downstaging success rates in patients with more or less advanced liver disease, although other studies have reported lower success in patients with Child’s C cirrhosis.65 Overall, the studies are variable in terms of success of downstaging, but overall it can be expected that approximately half of patients that are attempted to be downstaged will actually undergo LT.

Hepatic resection is the preferred curative treatment for patients with small localized tumors and well compensated liver disease and is an option for downstaging.68 Comprehensive pathological examination of resected specimens may facilitate the identification of patients with histological features of poor prognosis, for instance macrovascular invasion gone unobserved.69 This significantly influences subsequent treatment choices during postoperative surveillance of tumor recurrence patterns. Although large lesion size is not an absolute contraindication to hepatic resection, portal hypertension and end-stage liver disease are major risk factors for postsurgical complications and death.70 There is a subset of patients who require resection in conjunction with LRT to complete downstaging. However, surgical resection has been reported in a minority of studies as a downstaging modality so no statement can be made about its efficacy.

TACE is the most frequently used palliative treatment technique in downstaging protocols, particularly for multifocal HCC.43 The reported downstaging success rates with TACE (23.7–90%) are inconsistent and should be interpreted with caution.64 Since the TACE mechanism of action targets the hepatic arterial supply, its efficacy depends on responsive HCCs with good blood supply and uptake. While TACE is not advised to be performed in the presence of portal vein thrombosis, transarterial radioembolization with Yttrium-90 (Y-90) beads is a safe alternative downstaging therapy.71,72 Per available data there is no statistically significant difference between success rates of TACE and radioembolization for downstaging.65 It is important to note the risk of inaccurate staging when relying on imaging results to gauge radiological response to TACE or radioembolization in terms of tumor size and viability. For example, tumor response to Y-90 typically evolves gradually and may require 3–6 months to exhibit an adequate response on triphasic computed tomography (commonly known as CT) or magnetic resonance imaging.73 Therefore, timely intervals between treatment sessions and imaging are crucial to reduce confounding by image interpretation.

RFA confers its curative effects through thermal energy to achieve complete necrosis at a success rate of up to 90% in tumors of ≤3.0 cm in diameter.74 The rare complication of tumor seeding and risk of bleed with superficially located tumors are a few limitations within RFA’s safety profile.42 RFA is contraindicated near large vessels because of the heat sink effect, whereas MWA is a safe therapeutic option.70,75 SBRT, an extracorporeal technique, administers high doses of radiation to the target tumor. Published data investigating SBRT for downstaging are scant, but it appears to be a safe LRT for patients with decompensated liver function, especially in tumors near the major bile ducts.76,77

No evidence appears to render the superiority of one downstaging modality over another. The heterogeneity in the quality of data on the downstaging effectiveness of LRTs warrants large, multicenter, prospective cohort studies enriched with multidisciplinary tumor board referrals and standardized data reporting criteria in regions of differing waitlist times.

Systemic therapy and immunotherapy in advanced HCC

The goal of treatment is to maximize survival while prolonging the highest quality of life. Hence, it is paramount to assess the strength of scientific data for the selection of an appropriate treatment approach in HCC patients with advanced disease. When liver-directed therapy fails to successfully downstage patients into MC, HCC patients often transition into systemic therapy. Sorafenib is an oral tyrosine kinase inhibitor, whose anti-vascular endothelial growth factor (i.e. VEGF) receptor properties are proven to improve survival in advanced HCC patients, with a median survival of 10.7 months compared to a median survival of 7.9 months in placebo controls.78,79 In the scenario of sorafenib’s failure as a first-line systemic therapy, regorafenib, followed by cabozantinib, demonstrates a comparable survival benefit as second-line systemic therapy.80,81 Recently, in an open-label, phase III, multicenter, non-inferiority trial, lenvatinib, another oral multikinase inhibitor, displayed clinically meaningful improvement in objective response rate, progression-free survival, and time to progression compared to sorafenib in unresectable and treatment-naive HCC. However, the median survival was not statistically significantly between 13.6 months for lenvatinib and 12.3 months for sorafenib, (hazard ratio of 0.92, 95% confidence interval of 0.79–1.06).82 Newly, the REACH-2 phase III trial established the efficacy of ramucirumab, a monoclonal antibody that antagonizes VEGF receptor 2, in sorafenib-refractory patients with high AFP (of at least 400 ng/mL).83 Notwithstanding the emergence of systemic therapies, it is pertinent to mention that the role of the systemic therapies remains under study in the tumor downstaging to transplant setting. In a pilot, single-center, randomized controlled trial, the safety and adverse event profile of sorafenib plus Y-90 was compared to Y-90 alone in HCC patients as a bridge to LT. Data from the study’s limited sample size suggests the combination of sorafenib plus Y-90 in patients awaiting LT was linked with more peri-transplant biliary complications and a trend of higher acute cellular rejection rates.84 Given the lack of robust data, further studies are required to investigate and elucidate the utility of tyrosine kinase inhibitors or other systemic therapies in the pre-LT patient population, with regards to both efficacy and safety in the transplant setting. Tyrosine kinase inhibitors are known to inhibit wound healing, and patients who undergo liver transplant while being treated with tyrosine kinase inhibitors may be at risk for higher complications.

In cases of unresponsiveness or unfitness to receive tyrosine kinase inhibitors, negative regulators of T cell immune function, such as programmed cell death protein 1/programmed death ligand 1 (i.e. PD-1/PDL-1) or cytotoxic T-lymphocyte-associated antigen 4 (i.e. CTLA-4), have been identified as potential therapeutic targets.85 Two PD-1 checkpoint inhibitors, nivolumab and pembrolizumab, are promising immunotherapies for advanced HCC as second-line therapies.86,87 These two immunotherapies remain under Food and Drug Administration conditional approval, based on phase II data. There is also a recent approval of atezolizumab in combination with bevacizumab for treatment of advanced HCC, as well as the combination of PD-1 with CTLA-4 immunotherapy (nivolumab and ipilimumab). The lack of safety data with immunotherapy prior to transplant warrants further investigation. There is little to no data in the literature on the effects of immunotherapy in the liver transplant setting, with regards to the possibility of hyperacute or acute rejection after treatment.

AFP response to LRT

In the context of downstaging, the degree of a decrease in AFP in response to LRT is a valuable indicator of tumor biological aggressiveness. Policy implemented in the USA requires patients with AFP >1,000 ng/mL to exhibit a reduction in AFP to <500 ng/mL with LRT before proceeding with LT, in an effort to preserve comparable 5-year survival rates between HCC and non-HCC LT recipients.61,88 Recently, Mehta et al.89 endeavored to retrospectively validate the effects of this USA national policy using the UNOS database. In a multivariable analysis, a reduction in AFP from >1,000 ng/mL to 101–499 ng/mL was correlated with a greater than 2-fold reduction in post-LT death and close to a 3-fold reduction in HCC recurrence. The French AFP model identified a stricter AFP cutoff of ≤100 ng/mL for the subgroup of patients outside the MC as a predictor of nearly 70% 5-year overall survival rates and a low risk of recurrence.36 Interestingly, increasing AFP slope as low as 7.5 ng/mL/month and as high as 15 ng/mL/month in spite of LRT is associated with unfortunate outcomes in patients awaiting LT.90 While the implications of an AFP slope may seem irrelevant in world regions without a minimum 6 months waiting time, an observation period is essential for the “ablate and wait” strategy.91 Thus, the lack of durable response to LRT measured by AFP captures a supplementary exclusion criterion for LT.

18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)

Another potential diagnostic tool for patients being downstaged is 18F-FDG-PET imaging. Increased 18F-FDG accumulation of HCC consistently reflects tumor aggressiveness and is connected to undesirable post-LT survival.92,93 Poorly differentiated HCC expresses high 18F-FDG metabolism with a lesion-to-liver uptake ratio of more than 2.94 Despite its high sensitivity for detecting extrahepatic metastases, 18F-FDG-PET is not a widespread routine imaging modality due to the absence of cost-effectiveness analyses and prospective validation studies in regions with scarce donor resources.95 Ultimately, 18F-FDG-PET scans can help determine appropriate treatment options for 18F-FDG-PET-positive patients beyond MC by clarifying aggressiveness of disease.

Conclusions

In light of growing societal demands for LT, tumor downstaging surfaces at the heart of efforts to optimize the LT selection scheme. The premise of downstaging is to allow the opportunity of LT to a larger portion of HCC patients without affecting the transplant survival benefit. A multitude of robust data emphasize that the sole reliance on radiologic tumor size and number is a relatively crude method to gauge the complexity of HCC cases. Meanwhile, limited organ supply and waitlist life expectancy stress the value of surrogates for refined patient selection. AFP and novel biomarkers, LRT approaches, radiographic and AFP response to LRT, in combination with 18F-FDG-PET scans could be utilized as predictors of post-LT outcomes in a multifaceted LT evaluation process. Forthcoming longitudinal multicenter, well-designed studies are necessary to identify and prospectively validate reliable selection parameters. Overall, regional disparities in LT wait times and program-specific practices, like live donor LT, dictate patient eligibility for downstaging and individualized treatment decisions per recommendation and thorough follow-up by the program’s multidisciplinary team involving, but not exclusively, radiologists, hepatologists, surgeons, pathologists, and oncologists. Given the complexity of this disease, it is difficult to determine one particular downstaging method that is most successful, as each patient needs to be evaluated on an individual basis for which pre-LT treatment they can tolerate and will best downstage them to within transplant criteria. In general, careful patient selection combined with aggressive locoregional therapy appears to have the best outcomes long-term.

Abbreviations

18F-FDG: 

18F-fluorodeoxyglucose

AASLD: 

American Association for the Study of Liver Disease

AFP: 

alpha-fetoprotein

AFP-L3: 

lens culinaris agglutinin-reactive alpha-fetoprotein

CT: 

computed tomography

CTLA-4: 

cytotoxic T-lymphocyte-associated antigen 4

DCP: 

des-γ-carboxy prothrombin

EASL: 

European Association for the Study of the Liver

ETC: 

extended Toronto criteria

HCC: 

hepatocellular carcinoma

LT: 

liver transplantation

LRT: 

local-regional therapy

MC: 

Milan criteria

mRECIST: 

modified response evaluation criteria in solid tumors

MWA: 

microwave ablation

OLT: 

orthotopic liver transplantation

OPTN: 

Organ Procurement and Transplantation Network

PD-1/PDL-1: 

programmed cell death protein 1/programmed death ligand 1

PET: 

positron emission tomography

RFA: 

radiofrequency ablation

SBRT: 

stereotactic body radiotherapy

TACE: 

transarterial chemoembolization

UCSF: 

University of California in San Francisco

UNOS: 

United Network for Organ Sharing

VEGF: 

vascular endothelial growth factor

Y-90: 

Yttrium-90

Declarations

Data sharing statement

All data are available upon request.

Funding

None to declare.

Conflict of interest

Catherine Frenette reports the following conflicts of interest: Speakers Bureau for Bayer, Bristol Meyers Squibb, Eisai, Exelixis, Genentech; Advisory Board/Consultancy for Bayer, Eisai, Exelixis, Genentech; Research Support from Bayer, Merck, and Exelixis. The other author has no conflict of interests related to this publication.

Authors’ contributions

Writing and editing this manuscript (LF, CF).

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(6):394-424 View Article
  2. Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol 2019;16(10):589-604 View Article
  3. El-Serag HB. Hepatocellular carcinoma. N Engl J Med 2011;365(12):1118-1127 View Article
  4. Kim WR, Smith JM, Skeans MA, Schladt DP, Schnitzler MA, Edwards EB, et al. OPTN/SRTR 2012 Annual Data Report: liver. Am J Transplant 2014;14(Suppl 1):69-96 View Article
  5. Halazun KJ, Patzer RE, Rana AA, Verna EC, Griesemer AD, Parsons RF, et al. Standing the test of time: outcomes of a decade of prioritizing patients with hepatocellular carcinoma, results of the UNOS natural geographic experiment. Hepatology 2014;60(6):1957-1962 View Article
  6. El-Serag HB, Siegel AB, Davila JA, Shaib YH, Cayton-Woody M, McBride R, et al. Treatment and outcomes of treating of hepatocellular carcinoma among Medicare recipients in the United States: a population-based study. J Hepatol 2006;44(1):158-166 View Article
  7. Iwatsuki S, Starzl TE, Sheahan DG, Yokoyama I, Demetris AJ, Todo S, et al. Hepatic resection versus transplantation for hepatocellular carcinoma. Ann Surg 1991;214(3):221-228 View Article
  8. Ringe B, Pichlmayr R, Wittekind C, Tusch G. Surgical treatment of hepatocellular carcinoma: experience with liver resection and transplantation in 198 patients. World J Surg 1991;15(2):270-285 View Article
  9. Sotiropoulos GC, Molmenti EP, Lösch C, Beckebaum S, Broelsch CE, Lang H. Meta-analysis of tumor recurrence after liver transplantation for hepatocellular carcinoma based on 1,198 cases. Eur J Med Res 2007;12(10):527-534
  10. Zimmerman MA, Ghobrial RM, Tong MJ, Hiatt JR, Cameron AM, Hong J, et al. Recurrence of hepatocellular carcinoma following liver transplantation: a review of preoperative and postoperative prognostic indicators. Arch Surg 2008;143(2):182-188 View Article
  11. Welker MW, Bechstein WO, Zeuzem S, Trojan J. Recurrent hepatocellular carcinoma after liver transplantation - an emerging clinical challenge. Transpl Int 2013;26(2):109-118 View Article
  12. Rodríguez-Perálvarez M, Luong TV, Andreana L, Meyer T, Dhillon AP, Burroughs AK. A systematic review of microvascular invasion in hepatocellular carcinoma: diagnostic and prognostic variability. Ann Surg Oncol 2013;20(1):325-339 View Article
  13. Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, et al. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med 1996;334(11):693-699 View Article
  14. Mazzaferro V, Bhoori S, Sposito C, Bongini M, Langer M, Miceli R, et al. Milan criteria in liver transplantation for hepatocellular carcinoma: an evidence-based analysis of 15 years of experience. Liver Transpl 2011;17(Suppl 2):S44-S57 View Article
  15. Yao FY, Ferrell L, Bass NM, Bacchetti P, Ascher NL, Roberts JP. Liver transplantation for hepatocellular carcinoma: comparison of the proposed UCSF criteria with the Milan criteria and the Pittsburgh modified TNM criteria. Liver Transpl 2002;8(9):765-774 View Article
  16. Heimbach JK, Kulik LM, Finn RS, Sirlin CB, Abecassis MM, Roberts LR, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology 2018;67(1):358-380 View Article
  17. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182-236 View Article
  18. Yao FY, Ferrell L, Bass NM, Watson JJ, Bacchetti P, Venook A, et al. Liver transplantation for hepatocellular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology 2001;33(6):1394-1403 View Article
  19. Yao FY, Xiao L, Bass NM, Kerlan R, Ascher NL, Roberts JP. Liver transplantation for hepatocellular carcinoma: validation of the UCSF-expanded criteria based on preoperative imaging. Am J Transplant 2007;7(11):2587-2596 View Article
  20. Mazzaferro V, Llovet JM, Miceli R, Bhoori S, Schiavo M, Mariani L, et al. Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol 2009;10(1):35-43 View Article
  21. Toso C, Meeberg G, Hernandez-Alejandro R, Dufour JF, Marotta P, Majno P, et al. Total tumor volume and alpha-fetoprotein for selection of transplant candidates with hepatocellular carcinoma: A prospective validation. Hepatology 2015;62(1):158-165 View Article
  22. Zheng SS, Xu X, Wu J, Chen J, Wang WL, Zhang M, et al. Liver transplantation for hepatocellular carcinoma: Hangzhou experiences. Transplantation 2008;85(12):1726-1732 View Article
  23. Kaido T, Ogawa K, Mori A, Fujimoto Y, Ito T, Tomiyama K, et al. Usefulness of the Kyoto criteria as expanded selection criteria for liver transplantation for hepatocellular carcinoma. Surgery 2013;154(5):1053-1060 View Article
  24. Sapisochin G, Goldaracena N, Laurence JM, Dib M, Barbas A, Ghanekar A, et al. The extended Toronto criteria for liver transplantation in patients with hepatocellular carcinoma: A prospective validation study. Hepatology 2016;64(6):2077-2088 View Article
  25. Moher D, Dulberg CS, Wells GA. Statistical power, sample size, and their reporting in randomized controlled trials. JAMA 1994;272(2):122-124 View Article
  26. Euser AM, Zoccali C, Jager KJ, Dekker FW. Cohort studies: prospective versus retrospective. Nephron Clin Pract 2009;113(3):c214-c217 View Article
  27. Decaens T, Roudot-Thoraval F, Hadni-Bresson S, Meyer C, Gugenheim J, Durand F, et al. Impact of UCSF criteria according to pre- and post-OLT tumor features: analysis of 479 patients listed for HCC with a short waiting time. Liver Transpl 2006;12(12):1761-1769 View Article
  28. Clavien PA, Lesurtel M, Bossuyt PM, Gores GJ, Langer B, Perrier A. Recommendations for liver transplantation for hepatocellular carcinoma: an international consensus conference report. Lancet Oncol 2012;13(1):e11-e22 View Article
  29. Merani S, Majno P, Kneteman NM, Berney T, Morel P, Mentha G, et al. The impact of waiting list alpha-fetoprotein changes on the outcome of liver transplant for hepatocellular carcinoma. J Hepatol 2011;55(4):814-819 View Article
  30. Berry K, Ioannou GN. Serum alpha-fetoprotein level independently predicts posttransplant survival in patients with hepatocellular carcinoma. Liver Transpl 2013;19(6):634-645 View Article
  31. Harper AM, Edwards E, Washburn WK, Heimbach J. An early look at the Organ Procurement and Transplantation Network explant pathology form data. Liver Transpl 2016;22(6):757-764 View Article
  32. Bruix J, Reig M, Sherman M. Evidence-based diagnosis, staging, and treatment of patients with hepatocellular carcinoma. Gastroenterology 2016;150(4):835-853 View Article
  33. Toso C, Asthana S, Bigam DL, Shapiro AM, Kneteman NM. Reassessing selection criteria prior to liver transplantation for hepatocellular carcinoma utilizing the Scientific Registry of Transplant Recipients database. Hepatology 2009;49(3):832-838 View Article
  34. Hakeem AR, Young RS, Marangoni G, Lodge JP, Prasad KR. Systematic review: the prognostic role of alpha-fetoprotein following liver transplantation for hepatocellular carcinoma. Aliment Pharmacol Ther 2012;35(9):987-999 View Article
  35. Hameed B, Mehta N, Sapisochin G, Roberts JP, Yao FY. Alpha-fetoprotein level > 1000 ng/mL as an exclusion criterion for liver transplantation in patients with hepatocellular carcinoma meeting the Milan criteria. Liver Transpl 2014;20(8):945-951 View Article
  36. Duvoux C, Roudot-Thoraval F, Decaens T, Pessione F, Badran H, Piardi T, et al. Liver transplantation for hepatocellular carcinoma: a model including α-fetoprotein improves the performance of Milan criteria. Gastroenterology 2012;143(4):986-994.e3 View Article
  37. Lai Q, Avolio AW, Graziadei I, Otto G, Rossi M, Tisone G, et al. Alpha-fetoprotein and modified response evaluation criteria in solid tumors progression after locoregional therapy as predictors of hepatocellular cancer recurrence and death after transplantation. Liver Transpl 2013;19(10):1108-1118 View Article
  38. Sterling RK, Jeffers L, Gordon F, Venook AP, Reddy KR, Satomura S, et al. Utility of Lens culinaris agglutinin-reactive fraction of alpha-fetoprotein and des-gamma-carboxy prothrombin, alone or in combination, as biomarkers for hepatocellular carcinoma. Clin Gastroenterol Hepatol 2009;7(1):104-113 View Article
  39. Lok AS, Sterling RK, Everhart JE, Wright EC, Hoefs JC, Di Bisceglie AM, et al. Des-gamma-carboxy prothrombin and alpha-fetoprotein as biomarkers for the early detection of hepatocellular carcinoma. Gastroenterology 2010;138(2):493-502 View Article
  40. Chaiteerakij R, Zhang X, Addissie BD, Mohamed EA, Harmsen WS, Theobald PJ, et al. Combinations of biomarkers and Milan criteria for predicting hepatocellular carcinoma recurrence after liver transplantation. Liver Transpl 2015;21(5):599-606 View Article
  41. Halazun KJ, Najjar M, Abdelmessih RM, Samstein B, Griesemer AD, Guarrera JV, et al. Recurrence after liver transplantation for hepatocellular carcinoma: A new MORAL to the story. Ann Surg 2017;265(3):557-564 View Article
  42. Mazzaferro V, Battiston C, Perrone S, Pulvirenti A, Regalia E, Romito R, et al. Radiofrequency ablation of small hepatocellular carcinoma in cirrhotic patients awaiting liver transplantation: a prospective study. Ann Surg 2004;240(5):900-909 View Article
  43. Cescon M, Cucchetti A, Ravaioli M, Pinna AD. Hepatocellular carcinoma locoregional therapies for patients in the waiting list. Impact on transplantability and recurrence rate. J Hepatol 2013;58(3):609-618 View Article
  44. Yao FY, Bass NM, Nikolai B, Merriman R, Davern TJ, Kerlan R, et al. A follow-up analysis of the pattern and predictors of dropout from the waiting list for liver transplantation in patients with hepatocellular carcinoma: implications for the current organ allocation policy. Liver Transpl 2003;9(7):684-692 View Article
  45. Mehta N, Dodge JL, Goel A, Roberts JP, Hirose R, Yao FY. Identification of liver transplant candidates with hepatocellular carcinoma and a very low dropout risk: implications for the current organ allocation policy. Liver Transpl 2013;19(12):1343-1353 View Article
  46. Llovet JM, Bruix J, Fuster J, Castells A, Garcia-Valdecasas JC, Grande L, et al. Liver transplantation for small hepatocellular carcinoma: the tumor-node-metastasis classification does not have prognostic power. Hepatology 1998;27(6):1572-1577 View Article
  47. Bismuth H, Majno PE, Adam R. Liver transplantation for hepatocellular carcinoma. Semin Liver Dis 1999;19(3):311-322 View Article
  48. Park SJ, Freise CE, Hirose R, Kerlan RK, Yao FY, Roberts JP, et al. Risk factors for liver transplant waitlist dropout in patients with hepatocellular carcinoma. Clin Transplant 2012;26(4):E359-E364 View Article
  49. Llovet JM, Di Bisceglie AM, Bruix J, Kramer BS, Lencioni R, Zhu AX, et al. Design and endpoints of clinical trials in hepatocellular carcinoma. J Natl Cancer Inst 2008;100(10):698-711 View Article
  50. Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis 2010;30(1):52-60 View Article
  51. Samoylova ML, Dodge JL, Yao FY, Roberts JP. Time to transplantation as a predictor of hepatocellular carcinoma recurrence after liver transplantation. Liver Transpl 2014;20(8):937-944 View Article
  52. Heimbach JK, Hirose R, Stock PG, Schladt DP, Xiong H, Liu J, et al. Delayed hepatocellular carcinoma model for end-stage liver disease exception score improves disparity in access to liver transplant in the United States. Hepatology 2015;61(5):1643-1650 View Article
  53. Yao FY, Fidelman N. Reassessing the boundaries of liver transplantation for hepatocellular carcinoma: Where do we stand with tumor down-staging?. Hepatology 2016;63(3):1014-1025 View Article
  54. Majno PE, Adam R, Bismuth H, Castaing D, Ariche A, Krissat J, et al. Influence of preoperative transarterial lipiodol chemoembolization on resection and transplantation for hepatocellular carcinoma in patients with cirrhosis. Ann Surg 1997;226(6):688-701 View Article
  55. Thomas MB, Jaffe D, Choti MM, Belghiti J, Curley S, Fong Y, et al. Hepatocellular carcinoma: consensus recommendations of the National Cancer Institute Clinical Trials Planning Meeting. J Clin Oncol 2010;28(25):3994-4005 View Article
  56. Pomfret EA, Washburn K, Wald C, Nalesnik MA, Douglas D, et al. Report of a national conference on liver allocation in patients with hepatocellular carcinoma in the United States. Liver Transpl 2010;16(3):262-278 View Article
  57. Otto G, Herber S, Heise M, Lohse AW, Mönch C, Bittinger F, et al. Response to transarterial chemoembolization as a biological selection criterion for liver transplantation in hepatocellular carcinoma. Liver Transpl 2006;12(8):1260-1267 View Article
  58. Yao FY, Mehta N, Flemming J, Dodge J, Hameed B, Fix O, et al. Downstaging of hepatocellular cancer before liver transplant: long-term outcome compared to tumors within Milan criteria. Hepatology 2015;61(6):1968-1977 View Article
  59. Lai Q, Vitale A, Iesari S, Finkenstedt A, Mennini G, Spoletini G, et al. Intention-to-treat survival benefit of liver transplantation in patients with hepatocellular cancer. Hepatology 2017;66(6):1910-1919 View Article
  60. Mehta N, Guy J, Frenette CT, Dodge JL, Osorio RW, Minteer WB, et al. Excellent outcomes of liver transplantation following down-staging of hepatocellular carcinoma to within Milan criteria: A multicenter study. Clin Gastroenterol Hepatol 2018;16(6):955-964 View Article
  61. OPTN/UNOS Liver and Intestinal Organ Transplantation Committee. Changes to HCC criteria for auto approval. Available from: https://optn.transplant.hrsa.gov/media/1922/liver_hcc_criteria_for_auto_approval_20160815.pdf
  62. Mehta N, Dodge JL, Grab JD, Yao FY. National experience on down-staging of hepatocellular carcinoma before liver transplant: Influence of tumor burden, alpha-fetoprotein, and wait time. Hepatology 2020;71(3):943-954 View Article
  63. Samoylova ML, Nigrini MJ, Dodge JL, Roberts JP. Biases in the reporting of hepatocellular carcinoma tumor sizes on the liver transplant waiting list. Hepatology 2017;66(4):1144-1150 View Article
  64. Toso C, Mentha G, Kneteman NM, Majno P. The place of downstaging for hepatocellular carcinoma. J Hepatol 2010;52(6):930-936 View Article
  65. Parikh ND, Waljee AK, Singal AG. Downstaging hepatocellular carcinoma: A systematic review and pooled analysis. Liver Transpl 2015;21(9):1142-1152 View Article
  66. Barakat O, Wood RP, Ozaki CF, Ankoma-Sey V, Galati J, Skolkin M, et al. Morphological features of advanced hepatocellular carcinoma as a predictor of downstaging and liver transplantation: an intention-to-treat analysis. Liver Transpl 2010;16(3):289-299 View Article
  67. Ravaioli M, Grazi GL, Piscaglia F, Trevisani F, Cescon M, Ercolani G, et al. Liver transplantation for hepatocellular carcinoma: results of down-staging in patients initially outside the Milan selection criteria. Am J Transplant 2008;8(12):2547-2557 View Article
  68. Belghiti J. Resection and liver transplantation for HCC. J Gastroenterol 2009;44(Suppl 19):132-135 View Article
  69. Sala M, Fuster J, Llovet JM, Navasa M, Solé M, Varela M, et al. High pathological risk of recurrence after surgical resection for hepatocellular carcinoma: an indication for salvage liver transplantation. Liver Transpl 2004;10(10):1294-1300 View Article
  70. Llovet JM, Schwartz M, Mazzaferro V. Resection and liver transplantation for hepatocellular carcinoma. Semin Liver Dis 2005;25(2):181-200 View Article
  71. Iñarrairaegui M, Pardo F, Bilbao JI, Rotellar F, Benito A, D’Avola D, et al. Response to radioembolization with yttrium-90 resin microspheres may allow surgical treatment with curative intent and prolonged survival in previously unresectable hepatocellular carcinoma. Eur J Surg Oncol 2012;38(7):594-601 View Article
  72. Salem R, Lewandowski RJ, Kulik L, Wang E, Riaz A, Ryu RK, et al. Radioembolization results in longer time-to-progression and reduced toxicity compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology 2011;140(2):497-507.e2 View Article
  73. Riaz A, Awais R, Salem R. Side effects of yttrium-90 radioembolization. Front Oncol 2014;4:198 View Article
  74. Pompili M, Mirante VG, Rondinara G, Fassati LR, Piscaglia F, Agnes S, et al. Percutaneous ablation procedures in cirrhotic patients with hepatocellular carcinoma submitted to liver transplantation: Assessment of efficacy at explant analysis and of safety for tumor recurrence. Liver Transpl 2005;11(9):1117-1126 View Article
  75. Martin RC, Scoggins CR, McMasters KM. Safety and efficacy of microwave ablation of hepatic tumors: a prospective review of a 5-year experience. Ann Surg Oncol 2010;17(1):171-178 View Article
  76. Sandroussi C, Dawson LA, Lee M, Guindi M, Fischer S, Ghanekar A, et al. Radiotherapy as a bridge to liver transplantation for hepatocellular carcinoma. Transpl Int 2010;23(3):299-306 View Article
  77. Eriguchi T, Takeda A, Sanuki N, Oku Y, Aoki Y, Shigematsu N, et al. Acceptable toxicity after stereotactic body radiation therapy for liver tumors adjacent to the central biliary system. Int J Radiat Oncol Biol Phys 2013;85(4):1006-1011 View Article
  78. Llovet JM, Bruix J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology 2003;37(2):429-442 View Article
  79. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 2008;359(4):378-390 View Article
  80. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;389(10064):56-66 View Article
  81. Abou-Alfa GK, Meyer T, Cheng AL, El-Khoueiry AB, Rimassa L, Ryoo BY, et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N Engl J Med 2018;379(1):54-63 View Article
  82. Kudo M, Finn RS, Qin S, Han KH, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet 2018;391(10126):1163-1173 View Article
  83. Zhu AX, Kang YK, Yen CJ, Finn RS, Galle PR, Llovet JM, et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2019;20(2):282-296 View Article
  84. Kulik L, Vouche M, Koppe S, Lewandowski RJ, Mulcahy MF, Ganger D, et al. Prospective randomized pilot study of Y90+/-sorafenib as bridge to transplantation in hepatocellular carcinoma. J Hepatol 2014;61(2):309-317 View Article
  85. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 2017;169(7):1327-1341.e23 View Article
  86. Zhu AX, Finn RS, Edeline J, Cattan S, Ogasawara S, Palmer D, et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol 2018;19(7):940-952 View Article
  87. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017;389(10088):2492-2502 View Article
  88. Mazzaferro V, Sposito C, Zhou J, Pinna AD, De Carlis L, Fan J, et al. Metroticket 2.0 model for analysis of competing risks of death after liver transplantation for hepatocellular carcinoma. Gastroenterology 2018;154(1):128-139 View Article
  89. Mehta N, Dodge JL, Roberts JP, Hirose R, Yao FY. Alpha-fetoprotein decrease from > 1,000 to < 500 ng/mL in patients with hepatocellular carcinoma leads to improved posttransplant outcomes. Hepatology 2019;69(3):1193-1205 View Article
  90. Giard JM, Mehta N, Dodge JL, Roberts JP, Yao FY. Alpha-fetoprotein slope >7.5 ng/mL per month predicts microvascular invasion and tumor recurrence after liver transplantation for hepatocellular carcinoma. Transplantation 2018;102(5):816-822 View Article
  91. Roberts JP, Venook A, Kerlan R, Yao F. Hepatocellular carcinoma: Ablate and wait versus rapid transplantation. Liver Transpl 2010;16(8):925-929 View Article
  92. Lee JD, Yang WI, Park YN, Kim KS, Choi JS, Yun M, et al. Different glucose uptake and glycolytic mechanisms between hepatocellular carcinoma and intrahepatic mass-forming cholangiocarcinoma with increased (18)F-FDG uptake. J Nucl Med 2005;46(10):1753-1759
  93. Torizuka T, Tamaki N, Inokuma T, Magata Y, Sasayama S, Yonekura Y, et al. In vivo assessment of glucose metabolism in hepatocellular carcinoma with FDG-PET. J Nucl Med 1995;36(10):1811-1817
  94. Ho CL, Yu SC, Yeung DW. 11C-acetate PET imaging in hepatocellular carcinoma and other liver masses. J Nucl Med 2003;44(2):213-221
  95. Ho CL, Chen S, Yeung DW, Cheng TK. Dual-tracer PET/CT imaging in evaluation of metastatic hepatocellular carcinoma. J Nucl Med 2007;48(6):902-909 View Article
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Hepatocellular Carcinoma: Downstaging to Liver Transplantation as Curative Therapy

Leana Frankul, Catherine Frenette
  • Reset Zoom
  • Download TIFF