Topic Highlight Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Nov 21, 2013; 19(43): 7515-7530
Published online Nov 21, 2013. doi: 10.3748/wjg.v19.i43.7515
Bridging and downstaging treatments for hepatocellular carcinoma in patients on the waiting list for liver transplantation
Maurizio Pompili, Francesca Romana Ponziani, Department of Internal Medicine, Università Cattolica del Sacro Cuore, 8-00168 Rome, Italy
Roberto Iezzi, Department of Bioimaging and Radiological Sciences, Università Cattolica del Sacro Cuore, 8-00168 Rome, Italy
Alfonso Wolfango Avolio, Transplant Center, Catholic University, 1-00168 Rome, Italy
Giampiero Francica, Interventional Ultrasound Unit, Pinetagrande Hospital, 81030 Castelvolturno, Italy
Author contributions: Pompili M designed the study, wrote the manuscript, and revised the final version of the article; Francica G, Ponziani FR, Iezzi R and Avolio AW contributed to the literature search and writing the manuscript.
Correspondence to: Maurizio Pompili, MD, Department of Internal Medicine, Università Cattolica del Sacro Cuore, Largo A. Gemelli, 8-00168 Roma, Italy. mpompili@rm.unicatt.it
Telephone: +39-6-30154334 Fax: +39-6-35502775
Received: August 28, 2013
Revised: October 15, 2013
Accepted: October 17, 2013
Published online: November 21, 2013

Abstract

Several therapeutic procedures have been proposed as bridging treatments for patients with hepatocellular carcinoma (HCC) awaiting liver transplantation (LT). The most used treatments include transarterial chemoembolization and radiofrequency ablation. Surgical resection has also been successfully used as a bridging procedure, and LT should be considered a rescue treatment in patients with previous HCC resection who experience tumor recurrence or post-treatment severe decompensation of liver function. The aims of bridging treatments include decreasing the waiting list dropout rate before transplantation, reducing HCC recurrence after transplantation, and improving post-transplant overall survival. To date, no data from prospective randomized studies are available; however, for HCC patients listed for LT within the Milan criteria, prolonging the waiting time over 6-12 mo is a risk factor for tumor spread. Bridging treatments are useful in containing tumor progression and decreasing dropout. Furthermore, the response to pre-LT treatments may represent a surrogate marker of tumor biological aggressiveness and could therefore be evaluated to prioritize HCC candidates for LT. Lastly, although a definitive conclusion can not be reached, the experiences reported to date suggest a positive impact of these treatments on both tumor recurrence and post-transplant patient survival. Advanced HCC may be downstaged to achieve and maintain the current conventional criteria for inclusion in the waiting list for LT. Recent studies have demonstrated that successfully downstaged patients can achieve a 5-year survival rate comparable to that of patients meeting the conventional criteria without requiring downstaging.

Key Words: Hepatocellular carcinoma, Bridging treatment, Downstaging, Liver cirrhosis, Liver transplantation, Liver resection, Waiting list, Waiting time, Dropout rate

Core tip: The bridging treatments for patients with hepatocellular carcinoma within Milan criteria listed for liver transplantation are useful in decreasing dropout rate from the waiting list and the experiences reported to date suggest a positive impact on post-transplant tumor recurrence and patient survival. The response to treatments may represent a surrogate marker of tumor biological aggressiveness and could be evaluated to prioritize hepatocellular carcinoma candidates in the waiting list. Advanced hepatocellular carcinoma may be downstaged to achieve the current conventional criteria for inclusion in the waiting list and successfully downstaged patients can achieve an excellent 5-year survival rate.



INTRODUCTION

Liver transplantation (LT) is the treatment of choice for patients with unresectable hepatocellular carcinoma (HCC) complicating liver cirrhosis because it allows the cure of both the tumor and the underlying chronic liver disease. HCC classified within the so-called Milan criteria (MC) (1 nodule smaller than 5 cm or no more than 3 nodules smaller than 3 cm)[1] is recognized everywhere as the standard indication for LT. However, after admission to the waiting list for LT, HCC patients can experience tumor growth beyond the conventional transplant criteria. Indeed, there is a high cumulative probability of drop-out from the waiting list for HCC patients due to intrahepatic or extrahepatic tumor progression. This probability has been reported to range between 7% and 11% at 6 mo and to be approximately 38% at 12 mo following enrollment by two papers published at the end of the 1990s by Llovet et al[2] and Yao et al[3]. The probability has been correlated with tumor characteristics, geographic origin, and length of time waiting for LT[4-6].

Allocation policies for HCC patients awaiting LT remain controversial in the era of the model for end-stage liver disease (MELD) for the management of the LT waiting list. Different models have been developed to quantify the risk of death in neoplastic and non-neoplastic patients[7-11]. As the neoplastic risk assessment is not considered in MELD, patients with unresectable HCC with a neoplasm fulfilling the MC have been considered exceptions in the American allocation system. According to this rule, patients with T2-HCC fulfilling the MC (a single tumor of 2-5 cm or 2-3 tumors each < 3 cm) enter the waiting list with a MELD score equal to 22 and are therefore given priority over patients with less decompensated disease who enter the waiting list according to their laboratory MELD score (6 to 21). In addition, T2-HCC patients also receive incremental points for every 3 mo spent on the waiting list[12,13]. A similar approach has been implemented in other allocation systems[14,15]. The 22 threshold has been set to offer to HCC patients the same drop-out probability of patients without malignancy[16]. More detailed studies based on a dynamic prognosis parallelism have been published, and more complex allocation models aimed at balancing the risk of death in HCC and non-HCC patients have been proposed; however, they have not been applied in clinical practice[7,17-21]. According to these studies, LT candidates with HCC have dropout rates lower than non-HCC candidates, although the rate is similar to that of standard MELD candidates with a score of less than 21. Therefore, HCC patients appear to have an advantage in the current system, raising the question of whether a calculated continuous HCC priority score should be developed that also considers some biological features of the tumor such as the α-fetoprotein (AFP) value, size, and rate of growth[17,18]. Indeed, HCC patients with a high AFP level achieve acceptable LT outcomes if their AFP levels can be reduced with locoregional therapy during the waiting period[22,23]. Furthermore, an inadequate response to HCC bridging therapy was shown to be a strong predictor of dropout probability in three single-center Italian studies[10,14,24], whereas both the serum AFP level and the response to locoregional therapy were related to tumor recurrence and death in a retrospective international multicenter cohort study[25].

Lastly, the development of the survival-benefit approach, which proposes ranking priority according to the benefit in survival between standard care and LT rather than crude survival figures, changed the perspective of the outcome evaluation system[26-28]. The practice most widely used since 2005, which is in accordance with the United Network for Organ Sharing rules, gives HCC patients with unresectable T1 neoplasms (a single nodule smaller than 2 cm) the same priority as patients listed for non-neoplastic diseases.

In this scenario, several therapeutic procedures have been proposed and largely used in the past as bridging neo-adjuvant treatments for patients listed for LT with HCC within the MC[29]. The rationale for their use is the possible decrease of the waiting list drop-out rate before transplantation and of HCC recurrence after transplantation, which is less than 15% in patients with HCC within the MC undergoing LT without any prior tumor treatment[30]. These beneficial effects could also improve the overall survival of transplanted patients. Both surgical resection and locoregional therapies can be used not only as bridging procedures to LT in T2-HCC patients but also to downstage HCC patients who do not initially meet the conventional transplant criteria[31]. According to this approach, patients can be safely listed for LT if they can reach and maintain for an adequate follow-up period the MC or slightly expanded criteria such as the University of California San Francisco criteria (UCSF) (a single HCC ≤ 6.5 cm or ≤ 3 tumors with the largest being ≤ 4.5 cm and a total tumor burden ≤ 8 cm)[32] or the up-to-7 criteria [HCCs with 7 as the sum of the size of the largest tumor (in cm) and the number of tumors][33]. The aim of downstaging is to select HCC patients with reasonably low rates of tumor recurrence after LT among those who are initially excluded according to the current number-size criteria[34].

In this paper, we analyzed the indications and results of the various neo-adjuvant treatment modalities currently administered to HCC patients awaiting LT to avoid exceeding the MC while on the waiting list as well as those used to downstage patients who do not meet the conventional transplant criteria.

NEO-ADJUVANT BRIDGING PROCEDURES FOR PATIENTS WITH HEPATOCELLULAR CARCINOMA AWAITING LIVER TRANSPLANTATION
Surgical resection

Liver resection (LR) can be theoretically used as a first-line bridging procedure to LT. However, in most transplant centers, transarterial chemoembolization (TACE) and percutaneous ablation therapies are the preferred bridging therapies. The theoretical advantages of surgery in this setting are twofold. The first advantage is the best possible control of tumor growth, as TACE and percutaneous treatments do not always achieve complete tumor necrosis. The second advantage is the possibility of selecting patients in whom pathological analysis of the resected specimen shows features suggesting poor prognosis in terms of tumor recurrence, such as undifferentiated histotype, satellitosis, microvascular invasion, or capsular effraction, who should immediately undergo evaluation for LT[35]. However, compared to non-surgical therapies, the surgical bridging approach to patients listed for LT implies higher costs, entails more peri-procedural risks, can only be proposed in well-compensated patients without severe portal hypertension, and can make the ensuing LT technically more difficult, with a higher risk of post-operative complications[36].

Moreover, important issues regarding tumor resectability should also be considered. Single exophytic or at least subcapsular neoplasms are easier to resect than multiple neoplasms or those located adjacent to the hilum or vena cava[37,38]. Furthermore, a location in the left lobe represents a more favorable condition, and the progress achieved in the laparoscopic resection of the liver has reduced the number of HCC patients with an absolute indication for LT[39]. However, the combination of LR and LT over time appears to be a reasonable strategy; HCC patients within the MC who have preserved liver function can undergo LR, limiting LT as a rescue treatment in cases of tumor recurrence or liver function failure (salvage LT)[40]. This approach allows a consequent saving of grafts, which can then be more efficaciously transplanted in other patients, and is supported by the discrepancy between the limited donor pool and the enormous number of LT candidates. However, there are important differences in access to LT according to cadaveric organ availability, blood group of the recipient, implementation of a living donor program, and degree of donor-recipient matching[41-44].

Although during the initial experience with LR, the overall survival and disease survival rates of patients undergoing secondary LT after HCC resection were significantly lower (due to higher perioperative mortality unrelated to HCC) than those observed in cirrhotic patients with HCC undergoing primary LT[45], favorable results have more recently been reported by Belghiti et al[46]. They showed that postoperative course, complications, and the 3- and 5-year survival rates did not differ significantly between cirrhotic HCC patients undergoing primary LT or secondary LT after resection. Similarly favorable results for salvage LT have been subsequently reported by other groups in patients initially submitted to LR within the MC[15,47] or the UCSF criteria[48].

Salvage LT has been shown to be effective not only in the setting of deceased donor LT but also in the setting of living donor LT, particularly in Asian countries. Compared to deceased donor LT, the main advantage of living donor LT is the reduction of the waiting list time, whereas the main drawback is represented by the occurrence of severe life-threatening complications among donors in approximately 1% of cases[49]. Indeed, Hwang et al[50] have shown that the combination of prior recipient hepatectomy and a living donor liver graft is feasible and provides excellent long-term survival in treated patients, and their results have recently been confirmed by other groups[51,52].

Notably, the option of salvage LT cannot be offered to all patients initially treated by LR, primarily due to HCC recurrence overcoming the conventional LT criteria, age over 65 years at the time of recurrence, and the presence of comorbidities preventing the feasibility of LT. In a series reported by Poon et al[53], approximately 80% of patients were still eligible for salvage LT at the time of tumor recurrence. In a recent paper by Liu et al[48], among 71 patients with HCC recurrence within the UCSF criteria, salvage LT could be performed in 39 patients (54.9%). Compared to 180 HCC patients who underwent primary LT, patients treated with salvage LT for HCC recurrence showed greater intraoperative blood loss and required more blood transfusions; however, perioperative mortality, post-transplant complications, HCC recurrence rates, and overall survival did not differ significantly between the two groups.

Transarterial chemoembolization

TACE is considered the standard treatment for patients with intermediate-stage HCC according to the Barcelona-Clinic Liver Cancer classification[54], and it achieves a partial response in 15%-55% of patients and an improvement of median survival from 16 to 20 mo[55]. The most widely used conventional TACE procedure consists of an arterial infusion of a lipiodol emulsion with a chemotherapeutic agent (e.g., doxorubicin or cisplatin) followed by embolization with gelfoam. However, conventional TACE is not a standardized procedure, and the optimal chemotherapeutic/embolizing agent and retreatment strategy have yet to be determined[56]. In particular, it is well known that TACE requires treatment repetition either at regular intervals or ‘‘a la demande’’ and that repeating conventional TACE may damage non-cancerous hepatocyte functions and affect the clinical course. Indeed, liver toxicity is a major limitation of conventional TACE regimens, and superselective TACE is recommended in the setting of patients waiting for LT to minimize ischemic injury to non-tumoral liver tissue. Promising new data have been obtained using drug-eluting beads (DEBs), which are particles of variable size that are able to bind and elute doxorubicin in a predictable manner[57]. Compared to conventional techniques, DEBs appear to be a more standardized approach to TACE with less liver-related toxicity and fewer systemic adverse events[58].

TACE has been extensively used in the past as a bridging treatment to LT, and a number of studies have shown that it is an effective therapy in terms of adequate tumor necrosis achievement at explant analysis. Analyzing the largest available series indicates that the rate of patients treated by TACE reaching complete tumor necrosis is quite uniform, ranging between 27% and 57% in patients within the MC[59-67].

Interestingly, the rate of tumor necrosis appears to be higher in patients with single nodules when compared with patients with multiple nodules, in patients submitted to superselective TACE when compared with lobar TACE (complete necrosis achieved in 53.8% vs 29.8% of cases, respectively), and in patients with nodules 3-5 cm in size compared with patients with nodules smaller than 3 cm[65]. This last finding confirms the result obtained by Alba et al[64] and may be explained considering that larger nodules are typically fed by larger arteries, whereas in some instances, smaller nodules lack fully developed arterial neoangiogenesis; as a result, chemoembolization may be more effective in the former[65]. Accordingly, Kwan et al[66] have recently shown that the development of > 90% lesion necrosis upon pathological analysis of explanted liver was associated with avid lesion enhancement and the presence of a feeding vessel larger than 0.9 mm in diameter on the pre-TACE visceral angiogram. On post-TACE computed tomography images, a lack of residual contrast enhancement, a decrease in lesion size, a high lesion density due to an accumulation, and a diffuse distribution of ethiodized oil throughout the lesion were also correlated with near-complete lesion necrosis.

A recent small retrospective study compared tumor response in explanted liver after treatment with DEBs or standard TACE. TACE with DEBs achieved complete necrosis in 77% of the lesions, which was significantly higher than that reached after standard TACE (27.2%). More data are needed to address the better performance of DEBs compared to standard TACE in the transplant setting[68].

Another important point to clarify is the evaluation of TACE safety in patients awaiting LT. Because arteritis of the celiac and hepatic arteries may complicate TACE as a result of endovascular trauma caused by guides and catheters, recipients could be exposed after the transplant to an increased occurrence of complications such as arterial thrombosis. However, the prevalence of such serious complications has not been found to be increased in some studies comparing patients with or without TACE performed before LT[69-71].

Radiofrequency ablation

Radiofrequency thermal ablation (RFA) has gained widespread use over recent years as an effective procedure for small HCCs not amenable to surgical resection. Thermal ablation may be performed using cool-tip or hook needles with comparable results[72]. Some studies have described the use of RFA as a bridge to transplantation in HCC patients in recent years. These studies have reported complete tumor necrosis at pathological evaluation of the explanted liver in 47%-75% of cases, with a mean value of 58%[73-77]. A clear difference in effectiveness can be observed when analyzing tumors according to size. Indeed, the rate of complete necrosis ranges between 50% and 78% in HCCs up to 3 cm and between 13% and 43% in larger neoplasms[73-75,77]. Furthermore, in two studies, a tumor size larger than 3 cm was the only risk factor identified for HCC persistence after treatment[73,75].

Regarding RFA-related complications in the setting of HCC patients awaiting LT, an analysis of the largest available series demonstrated that the procedure is quite safe. In fact, considering 5 large series, the mean rate of post-ablation major complications was only as high as 4.6%, including one case of death due to peritoneal bleeding, two cases of acute peritonitis/cholecystitis, and one case each of severe liver failure treated by urgent transplantation, severe persistent liver failure, biliary stenosis, arterial hemorrhage, and small bowel perforation[73-76,78]. Additionally, the risk of tumor seeding at the level of the abdomen wall appears to be low; however, occasional cases of tumor seeding along the needle track diagnosed after LT in patients submitted to RFA as a bridging procedure have been reported in the literature[79,80].

Other treatments

TACE and RFA are the most used bridging treatments to LT in HCC patients, although other therapeutic options have been proposed (Table 1). Percutaneous ethanol injection (PEI) is the oldest and most used technique for the local treatment of HCC, but it has been rarely used as a bridging treatment to transplantation. In our multicenter survey, the rate of complete necrosis in tumors smaller than 3 cm was 30%[75]. Castroagudín et al[81], in a series of 20 nodules in 19 patients, showed that in patients with small tumors (i.e., less than 3 cm), ethanol injection induced complete necrosis in 58% of the cases. In a more recent paper, Branco et al[82] reported a complete necrosis rate of 64% in 59 patients within the MC and a mean tumor size of 2.4 cm (range: 0.5-5.5 cm). In these studies, PEI was not affected by procedure-related major complications and did not provide total necrosis in most tumors larger than 3 cm.

Table 1 Advantages and disadvantages of the bridging and downstaging procedures for hepatocellular carcinoma in cirrhotic patients who are candidates for liver transplantation.
AdvantagesDisadvantages
ResectionHigher complete effectiveness than non-surgical proceduresUnfeasible in patients with decompensated liver disease or severe portal hypertension
More simple in cases with peripheral subglissonian nodules
TACEMore effective using the selective/superselective technique in well-vascularized nodules with large feeding arteriesUnfeasible in patients with severely reduced portal vein flow, intratumoral arteriovenous fistula, or renal failure (creatinine clearance < 30 mL/min)
TAREPossible better effectiveness than TACE in cases with multiple nodulesLess experience with TARE than TACEHigh cost
RFAMore effective in nodules ≤ 3 cmPotentially dangerous in patients with impaired clotting parameters or lesions located superficially or near the gallbladder, major bile ducts, or bowel loops
PEIMore effective in nodules ≤ 3 cmSuitable in patients with impaired clotting parameters or lesions located in dangerous sites for thermal ablationLess effective than RFA for nodules > 2 cm
PLAMore effective in nodules ≤ 3 cmSuitable in patients with impaired clotting parametersLess experience with PLA than RFATechnically complexPotentially dangerous in cases of lesions located superficially or near the gallbladder, major bile ducts, or bowel loops
MWAPossible better effectiveness than RFA in nodules ≥ 3 cm or located near large vesselsLess experience with MWA than RFAPotentially dangerous in patients with impaired clotting parameters or withlesions located superficially or near the gallbladder, major bile ducts, or bowel loops

Percutaneous laser ablation (PLA) performed using multiple tiny laser fibers has recently been shown to be an effective technique for the thermal ablation of HCC in patients in whom surgical resection is not possible or appropriate[83,84]. We recently showed that in HCC patients awaiting LT, PLA provided results comparable to those of RFA; the rate of complete necrosis found at explant analysis in a series of 13 nodules up to 3 cm was 62%[85]. Due to the use of fine needles, the possible advantages of PLA in respect to RFA include the treatment of patients with either nodules in high-risk sites (i.e., near vital structures)[86] or severe clotting impairment, in whom RFA may be contraindicated, and the lower overall cost of the procedure.

Microwave ablation (MWA) has been shown to be an effective thermal ablation procedure for the percutaneous treatment of HCC. Compared to RFA, this technique could theoretically provide a larger volume of necrosis and be more effective when treating nodules adjacent to large vessels; however, a clear advantage of MWA with respect to RFA has not been demonstrated[87,88]. The use of MWA as a bridging procedure to LT or a downstaging procedure in HCC patients appears to be promising. In a recent preliminary study, 6 patients with 6 HCC nodules ranging between 2.5 and 5.0 cm (mean 3.5 cm) in diameter underwent MWA before LT. At explant analysis, all of the nodules showed complete necrosis without intraoperative evidence of tumor spread in all cases or evidence of tumor recurrence at a one-year follow up in the 5 patients who could be evaluated[89].

The effectiveness of transarterial radioembolization (TARE) with 90Yttrium microspheres has recently been evaluated by Riaz et al[90], who studied 38 nodules in 35 patients. Of 15 patients with T2-HCC, none progressed to T3-HCC (one nodule > 5 cm or up to three nodules with one > 3 cm) before LT, whereas 8 of 10 patients were downstaged from stage T3 to stage T2. At explant analysis, 23 of the 38 target lesions (61%) showed complete tumor necrosis, and its achievement was affected by the size of the target lesion; indeed, complete necrosis was detected in 89%, 65%, and 33% of lesions smaller than 3 cm, between 3 and 5 cm, and larger than 5 cm, respectively.

Data regarding the use of external conformal radiotherapy (CRT) as a bridging treatment to LT in HCC patients are scarce. In a recent paper, CRT was delivered in five or six fractions to 10 patients with HCC awaiting LT with tumor diameters ranging from 2.5 to 10.8 cm. Nine patients completed the treatment, and it was well tolerated in all cases. Two tumors remained stable; the rest had 10%-50% regression, which was sustained on follow-up imaging. Five patients underwent LT, and at explant pathology, tumor necrosis ranging between 40% and 90% was demonstrated. No patients showed tumor recurrence after LT (median follow-up period of 6 mo). The main conclusions of the paper were that CRT is a safe and efficacious local bridging therapy for patients with HCC who are on the waiting list for LT and that further studies are warranted to compare the effectiveness of CRT to other local treatment regimens[91].

Combined treatments

Experiences with combined therapies such as TACE followed by RFA[92-94] or RFA shortly after TACE[95] have been published in recent years, typically in the setting of unresectable HCC larger than 3 cm. The rationale for the use of combined treatment rather than a single treatment is to reach a higher local tumor control rate due to higher rates of complete tumor necrosis. In this context, the question arises of how TACE and RFA should be sequenced. The advantage of performing TACE prior to RFA is the reduced heat-sink effect with the ability to create larger ablation zones more easily. The advantage of using TACE after RFA is that RFA generates a hyperemic rim surrounding the ablation area, which can consequently be targeted by transarterial means more effectively. The approach of combined treatment may be applied even as a bridge to LT. A recent experience with combined TACE followed by RFA in a series of 44 HCC patients within the MC reported the absence of major complications and a 76.9% rate of complete necrosis in the 16 patients with 26 nodules who underwent LT[96].

IMPACT OF BRIDGING TREATMENTS ON DROPOUT FROM THE LIVER TRANSPLANTATION WAITING LIST

The impact of bridging treatments on waiting list dropout is uncertain due to the absence of prospective comparative studies, but the dropout data of treated patients should be compared with the features of HCC patients awaiting LT without any bridging treatment. In the latter case, the dropout rates were greater than 30% 12 mo after being added to the list[2,3]. In 2006, Lesurtel et al[97] published an interesting paper dealing with the usefulness of TACE in HCC patients undergoing LT according to the criteria of evidence-based medicine. The question was whether TACE impacted the waiting list dropout rate. They found insufficient evidence to answer this question. Hayashi et al[61] reported a discouraging 35% dropout rate in patients with TNM stage 1 or 2 HCC and a mean waiting time of 340 d after treatment with TACE. Similarly, among 54 listed HCC patients who underwent TACE prior to LT, Maddala et al[98] revealed drop-out rates of 15% and 25% at 6 and 12 mo, respectively. However, the most recent series including patients treated with TACE before LT have indicated that the dropout rate due to tumor progression is lower and ranges between 3.0% and 9.3%, with a mean waiting time on the transplantation list exceeding 6 mo in the largest available studies[63,64,69] (Table 2).

Table 2 Selected studies on non-surgical bridging therapy for hepatocellular carcinoma before liver transplantation n (%).
Ref.TreatmentPatientsHCC stageDropout rate -Total -HCC progressionHCC recurrence after LTIntention-to-treat survivalSurvival after LT
Fontana et al[99]RFA33 (15 LT)MC (30 pts)NA2 (13)NA85% at 3 yr
Graziadei et al[60]TACE48 (41 LT)MC01 (2.4)94% at 5 yr94% at 5 yr
Hayashi et al[61]TACE20 (12 LT)MC6 (35)NA61% at 3 yr100% at 4 yr
Maddala et al[98]TACE54 (46 LT)MC (47 pts)8 (14.8)5 (13.3)61% at 5 yr74% at 5 yr
6 (11.1)
Mazzaferro et al[73]RFA50 (50 LT)MC (40 pts)0 (0)2 (4)NA83% at 3 yr
Lu et al[74]RFA52 (41 LT)MC (42 pts)6 (12)0 (0)74% at 3 yr76% at 3 yr
3 (5.8)
Castrogaudin et al[81]PEI34 (23 LT)UNOS T1-T2 (30 pts)5 (14.7)1 (4.3)NA19/23 (82.6%) alive (median FU 21 mo)
2 (5.9)
Pompili et al[75]RFA, PEI40 (40 LT)MC (37 pts)NA3 (7.5)NA85.4% at 3 yr
Porrett et al[79]TACE, RFA, TARE31 (31 LT)UNOS T1-T2NA7 (22.6)NA84% at 3 yr
Brillet et al[76]RFA21 (16 LT)MC5 (23.8)1 (6.3)NA11/16 (69%) alive (median FU 25 mo)
3 (14.3)
Millonig et al[63]TACE68 (66 LT)MC2 (3)5 (7.6)70% at 5 yrNA
Majno et al[69]TACE43 (43 LT)MC12 (27.9)4 (9.3)NANA
4 (9.3)
Rodríguez-Sanjuán et al[77]RFA28 (28 LT)MC (25 pts)NA2 (7.1)NANA
Alba et al[64]TACE63 (56 LT)MC7 (11)6 (10.7)NA60.4% at 5 yr
3 (4.8)
Branco et al[82]PEI62 (59 LT)MC3 (4.8)3 (5.1)64.4% at 3 yr67.7% at 3 yr
DuBay et al[78]RFA77 (51 LT)MC19 (25)1 (2)NA> 80% at 3 yr
16 (21)
Ashoori et al[96]TACE + RFA36 (16 LT)MC6 (16.7)0 (0)NA11/16 alive (median FU 29.9 mo)
4 (11.1)
Tsochatzis et al[67]TACE, TAE67 (67 LT)MCNA4 (6)NANA

Less data are available in this setting for patients submitted to RFA. In a preliminary study published in 2002, Fontana et al[99] reported a dropout rate of 21% over a mean waiting period of 7.9 mo among 33 patients treated with RFA prior to LT. In more recent papers including larger numbers of patients, the dropout rate due to HCC progression was found to be 0% after a mean waiting time of 9.5 mo in one study[73] and 5.8% at 12 mo in another study[74] (Table 2). In a large study including only HCC patients within the MC awaiting LT, 77 patients who underwent RFA were compared to 93 patients without any bridging treatment; a non-specific trend toward a higher dropout rate for tumor-specific events was detected among RFA patients (21% vs 11%), but the mean waiting time was significantly higher in the RFA group. Using survival analysis modeling, there was no significant difference in the time to dropout between the RFA and no-treatment groups for all causes[78].

Encouraging data have been reported following the application of multimodal schedules of treatment; in a series of 44 listed HCC patients within the MC who systematically underwent TACE followed by RFA, the intention-to-treat cumulative dropout rates were 5.5% and 11.0% at 12 and 24 mo, respectively[96].

Lastly, the short-term response to bridging treatment has recently been reported to be crucial in the prediction of dropout. In a recent report by De Giorgio et al[24], 170 HCC patients awaiting LT within the MC who underwent percutaneous ablation, TACE, or surgery as a bridging treatment were analyzed. Total tumor diameter and recurrence or persistence of tumor activity at the 6-wk follow-up after therapy were significantly correlated with progression beyond the MC and dropout from the waiting list. The finding of a significantly decreased dropout probability among T2 patients achieving a complete or partial response to bridging treatment compared with patients with an inadequate or no response to treatment has also been confirmed in two other large studies[10,14].

In summary, there is sufficient evidence to conclude that bridging treatments yielding complete or subtotal HCC necrosis on imaging effectively reduce the rate of dropout from the waiting list.

IMPACT OF BRIDGING TREATMENTS ON RECURRENCE OF HEPATOCELLULAR CARCINOMA AFTER LIVER TRANSPLANTATION

A less than 15% recurrence rate has been reported for HCC in patients within the MC undergoing LT without any treatment[30]. Whether the application of bridging therapies while on the waiting list decreases this rate is controversial. Again, prospective multicenter comparative studies are lacking in this field, and the only available data were obtained in single-center retrospective case series.

Regarding TACE, in a cohort of 111 HCC patients undergoing LT (54 treated preoperatively with TACE), Majno et al[59] showed that downstaging of tumors > 3 cm and total necrosis of the nodule at explant analysis were associated with better 5-year disease-free survival than either an inadequate response to TACE or no TACE before LT. Thereafter, low recurrence rates of 7.6% and 10.7% were reported in two large series of HCC patients within the MC who were treated with TACE before LT[63,64]. A clear trend toward longer recurrence-free survival has also been observed by Milllonig et al[63] in patients with complete tumor necrosis when compared with patients with viable tumor at explant analysis. More recently, Tsochatzis et al[67] evaluated 150 consecutive patients with HCC within the MC who underwent LT. Sixty-seven patients (45%) underwent transarterial embolization (TAE) with polyvinyl alcohol particles or TACE before LT, and the remaining 83 patients were not treated before LT. HCC recurrence after LT was significantly lower in the TAE-TACE group (6%) than in the no TAE-TACE group (18.1%) (Table 2). Furthermore, post-transplant HCC recurrence was independently associated with no neo-adjuvant transarterial therapy and the total radiological size of the HCC nodules.

HCC recurrence after LT has been evaluated in 7 studies of patients who underwent RFA as the only bridging treatment. In total, 231 patients were evaluated over follow-up periods of 15-41 mo (mean 28 mo). Overall, HCC recurrence was detected in 8 patients (3.5%), and the rate of recurrence ranged between 0% and 13%[73-78,99] (Table 2).

Some recent single-center studies appear to confirm a positive impact of bridging treatments on HCC recurrence after LT. In a series of 147 HCC candidates (38% outside the MC) who underwent RFA, TACE, or multimodal treatment before LT, a complete or partial response was observed in 57.8% of cases. Transplanted patients with stable disease or no response to pre-LT HCC treatment had a significant 6-fold increase in tumor recurrence after LT compared with patients with a complete or partial response (13% vs 2%)[10]. In another study that included 315 HCC patients who were candidates for LT (17% outside the MC) and underwent TACE, RFA, PEI, or surgical resection, a complete response to treatment was observed in 49.1% of cases; transplanted patients with a partial or no response to bridging treatments showed a significantly higher risk of HCC recurrence compared with patients with a complete response (19.4% vs 5.5%)[14]. Among 137 transplanted patients (42 outside the MC) who underwent locoregional bridging treatments such as resection, TACE, RFA, and PEI before LT, AFP > 400 ng/mL was the only significant pre-transplant factor linked to HCC recurrence after LT. Conversely, the use of locoregional treatments was a significant protective factor, and the best 5-year tumor-free survival was observed in patients within the MC who underwent locoregional treatment[100]. Lastly, within a group of 93 consecutive HCC patients (36 beyond MC) who underwent LT, 59 underwent pre-transplant TACE or RFA. The 5-year tumor-free survival did not significantly differ between treated and untreated patients (78% vs 68%). However, among the treated patients, the presence of more than 50% necrosis of the target lesions at explant analysis was associated with a significantly better 5-year tumor-free survival rate (96% vs 21%)[101].

Overall, a trend toward a decreased recurrence rate after LT appears to emerge in patients achieving a total or subtotal response to the treatment administered before LT.

IMPACT OF BRIDGING TREATMENTS ON SURVIVAL AFTER LIVER TRANSPLANTATION

Independent of the treatment administered, a key question remains to be answered: do bridging treatments improve survival in HCC patients who undergo LT? There is insufficient evidence of a beneficial effect of TACE because data obtained from prospective randomized studies are lacking[97]. A multicenter retrospective case control study from France compared 100 HCC patients who underwent TACE before transplantation and 100 HCC patients transplanted without any prior treatment. The 5-year survival (59% in both groups) and 5-year disease free survival (69% vs 64%) rates were not significantly different. At explant analysis, greater than 80% total or subtotal HCC necrosis was found in 30% of treated patients, and this subgroup showed a non-significant trend toward a better 5-year survival compared with a matched untreated control group (63% vs 54%)[62]. It can be reasonably argued that patients with total/subtotal tumor necrosis might receive a significant survival benefit from TACE before LT, perhaps due to a decreased risk of post-transplant HCC recurrence. This hypothesis appears to have been confirmed in a study by Millonig et al[63] that included 116 HCC patients who underwent TACE before transplantation. Most of the patients were within the MC, and complete tumor necrosis was found in 27% of the cases. The 5-year survival rate was higher in patients with completely necrotic tumors than in patients with partial necrosis (86% vs 66%), although this difference did not reach statistical significance.

The influence of neo-adjuvant treatments on post-LT survival should be analyzed independent of the treatment used; however, the only available data come from single-center retrospective series and provide contradictory results. In a study by Bharat et al[102], 46 HCC patients undergoing various bridging treatments before LT were compared to 46 matched HCC patients transplanted without any treatment. The 5-year survival rate was significantly higher in the treated group (82% vs 52%), although the survival advantage was evident only for patients with T2-T4 tumors, not for patients with T0-T1 tumors. Even the 5-year disease-free survival rate was slightly higher in the treated group (84% vs 76%), although this difference was not significant. In a study by Lao et al[103], 91 untreated HCC patients who underwent LT were compared to 33 patients with HCC who underwent TACE, RFA, or PEI before LT. HCC recurred only in 9 untreated patients, and the only factors significantly linked to tumor recurrence were a MELD score < 14, AFP > 1000 ng/mL, and the absence of pre-LT bridging treatment. The disease-free survival showed a non-significant trend toward a better outcome in treated patients, whereas the cumulative survival did not differ. Heckman et al[104] compared the outcomes of 50 HCC patients undergoing bridging therapy before LT to those of 73 HCC patients transplanted without any prior treatment; they found a non-significant trend toward improved 5-year survival in treated patients (81% vs 71%). Porrett et al[79] compared 30 treated patients to 33 untreated patients before transplant. Their study failed to show any survival difference between the groups, but it should be noted that only 20% of the treated patients had complete HCC necrosis at explant analysis. Lastly, in the previously cited study by DuBay et al[78], no differences in 5-year overall or tumor-free survival from the list date or transplant were identified when comparing 77 patients treated with RFA to 93 matched untreated patients. No data were provided about the achievement of complete necrosis in the ablated tumors at explant analysis.

Although a definitive conclusion cannot be made, a positive impact of pre-LT treatments on post-LT survival could be present. Indeed, in the United States, data on liver transplant activity for HCC from 1997 to 2006 demonstrated a higher 3-year post-LT survival in patients who underwent ablative procedures compared with patients who did not[105]. Moreover, studies reporting no difference between treated and untreated patients also tend to report shorter waiting times for LT[29].

DOWNSTAGING OF HCC BEYOND THE CONVENTIONAL LIVER TRANSPLANTATION CRITERIA

Downstaging of HCC to within the MC or the UCSF criteria is an attractive alternative to expanding the tumor size limits for LT. Theoretically, the downstaging process allows the selection of tumors with a more favorable biology that will likely respond to downstaging treatments and will also do well following LT[106].

In recent years, several papers have been published defining successful downstaging as fulfilling the MC[107-114] or the UCSF criteria[106]. However, different criteria for successful treatment have been used in other studies, including fulfilling the MC without a serum AFP level higher than 400 ng/mL[115], a 30%-50% decrease in the size of treated nodules[60,116], or no tumor progression during the downstaging treatment in patients with well or moderately differentiated HCC[4,117] (Table 3). In some of these studies, only TACE[60,108,110,112-114] or transarterial chemoinfusion[107] was used as the downstaging procedure, whereas in other studies, a multimodal approach was used, including TACE, RFA, PEI, or surgical resection[4,106,114,115,117]. TARE as a single downstaging procedure was retrospectively compared to TACE in a study by Lewandowski et al[109]. Better performance was observed for TARE in terms of the downstaging success rate and 3-year intention-to-treat post-HCC treatment survival.

Table 3 Selected studies on downstaging therapy for hepatocellular carcinoma before liver transplantation n (%).
Ref.TreatmentPtsInclusion criteria1Successful downstage-Criteria-RateTransplanted ptsRecurrence free survival after LTIntention to treat survivalSurvival after LT
Graziadei et al[60]TACE36HCC > 5 cmDecreased size > 50%11/36 (31)10Recurrent HCC: 3 pts (30)31% at 5 yr41% at 4 yr
Otto et al[116]TACE62Beyond MCDecreased size ≥ 30%34/62 (55)2768% at 5 yrNA73.2% at 5 yr
Cillo et al[4]TACE, RFA, PEI, Resection40Beyond MCWD or MD HCCMaintenance of selection criteriaNA31Recurrent HCC: 0 pts79% at 5 yr> 90% at 3 yr
Chapman et al[108]TACE76Beyond MCMC18/76 (24)1750% at 5 yrNA93.8% at 5 yr
Yao et al[106]TACE, RFA, Resection611 HCC 5-8 cm2-3 HCCs 3-5 cm, total diameter ≤ 8 cm4-5 HCCs ≤ 3 cm total diameter ≤ 8 cmUCSF43/61 (71)3592% at 2 yr69% at 4 yr92% at 2 yr
Ravaioli et al[115]Multimodal (TACE, PEI, RFA, Resection)481 HCC 5-8 cm2 HCCs 3-5 cm, total diameter ≤ 8 cm3-5 HCCs ≤ 4 cm total diameter ≤ 12 cmMC and AFP < 400 ng/mL32/48 (67)3271% at 3 yr62% at 3 yrNA
Lewandowski et al[109]TACE (43 patients)TARE (43 patients)86UNOS T3MCTACE 11/35 (31)TARE 25/43 (58)TACE 11TARE 9TACE 73% at 1 yrTARE 89% at 1 yrTACE 19% at 3 yrTARE 59% at 3 yrNA
De Luna et al[107]TACI27Beyond MCMC17/27 (63)15NA84.1% at 3 yr78.8% at 3 yr
Jang et al[110]TACE386Beyond MCMC or complete tumor necrosis160/386 (41.5)3766.3% at 5 yrNA54.6% at 5 yr
Barakat et al[111]TACE, TARE, RFA, Resection32Beyond UCSF (18 pts)Beyond MC (14 pts)UNOS T218/32 (56.3)13Recurrent HCC: 2 pts (15.4%)NA75% at 2 yr
Bargellini et al[112]TACE33Beyond MCComplete or partial response, or stable disease according to mRECIST criteriaNA3374.4% at 5 yrNA72.5% at 5 yr
Bova et al[113]TACE, TAE48Beyond MCMCAFP < 100 ng/mL19/48 (39)9Recurrent HCC: 1 pt (11.1%)NANA
Lei et al[114]TACE, RFA, Resection, HIFU58Beyond MC Within UCSFMCNA5863.8% at 5 yrNA74.1% at 5 yr

Significant factors for unsuccessful downstaging related to biological tumor features have been reported by some of these papers. In the study by Yao et al[106], AFP > 1000 ng/mL was the only significant negative prognostic factor. Barakat et al[111] showed that the mean AFP level and the rate of infiltrative tumors were significantly higher in patients who did not achieve successful downstaging. An AFP level lower than 100 ng/mL and the 3-year survival probability calculated using the Metroticket calculator[33] were the only independent predictors of successful downstaging in the study by Bova et al[113]. An AFP slope > 15 ng/mL per month and tumor progression according to the Modified Response Evaluation Criteria in Solid Tumors (mRECIST)[118] were independent risk factors for HCC recurrence and patient death in an international retrospective multicenter European study performed by Lai et al[25] that included MC-within (316 cases) and MC-outside (116 cases) patients who underwent LT after locoregional therapy. We should also highlight that after successful downstaging, some authors have recommended that patients undergo a 3-mo observation period before listing to assess the stability of neoplastic disease[4,106,115]. This “test of time” will identify rapidly recurring lesions, vascular invasion, and distant metastasis, thereby decreasing the risk of tumor recurrence and poor overall results after LT[34].

Overall, according to the presently available data, the successful downstaging rate ranges between 24% and 71% (Table 3). The proportion of patients transplanted ranges between 10% and 67%, and the average waiting time to LT ranges between 2 and 10.9 mo[29]. Additionally, the reported survival rates range from 78.8% to more than 90% and from 54.6% to 93.8% at 3 and 5 years, respectively[119]. Two prospective studies have demonstrated that survival after LT in patients with large tumors successfully downstaged within the MC[115] or the UCSF criteria[106] is similar to that of patients who initially met the criteria for transplantation. Six studies[4,60,107,108,114,116] compared patients who were downstaged successfully within the MC with those who initially met the MC. Five of these studies[4,107,108,114,116] reported no significant difference in absolute or disease-free survival between groups, whereas one study[60] reported that patients who were downstaged successfully had significantly worse survival at 1, 2, and 5 years after LT. Lastly, in a recent study, no significant differences in postoperative complications, tumor recurrence, or survival rate were reported between two groups of patients with advanced HCC who underwent deceased donor LT (52 patients) or living donor LT (31 patients) after successful downstaging therapy[42].

CONCLUSION

Currently, locoregional therapies play a crucial role in the treatment of patients awaiting LT. For patients listed within the MC (stage T2-HCC), a delay of LT over 6-12 mo without bridging treatment is a well-recognized risk factor for tumor progression and dropout from the list or interval dissemination with post-transplant tumor recurrence[2,3,16]. For this reason, the optimal strategy for T2-HCC patients awaiting LT should be to transplant within 6 mo without pre-transplant therapy[120]. However, if a longer waiting time is needed, following the current guidelines of the American Association for the Study of Liver Disease and the European Association for the Study of the Liver for the treatment of HCC[121,122] and the recommendations of a recent international consensus conference on the management of HCC patients who are LT candidates[123], the use of bridging treatments is recommended, as several studies in recent years have documented their usefulness in preventing tumor progression. There is, however, no evidence that bridging treatments are useful in patients with T1-HCC[123].

In patients who underwent previous liver resection and experienced tumor recurrence but are within the currently accepted transplant criteria or those with liver function failure, salvage LT using deceased donor livers yields an acceptable long-term survival rate and can be considered[6,25]. Salvage LT using living donors has also been successfully performed in centers with high-volume living donor programs, and they appear to provide long-term results comparable to those obtained using deceased donor grafts[48].

Regarding non-surgical bridging therapies, no recommendation can be made for one type of locoregional therapy over others[123]. However, RFA could be the first-line treatment for lesions up to 3 cm, in which complete tumor necrosis has been shown in more than 50% of cases at explant analysis. The risk of major complications related to RFA in this patient setting appears to be quite low, but it is good clinical practice to limit needle insertions and to avoid the treatment of superficially located lesions. PEI appears to show lower efficacy and can be reserved for small lesions located in sites considered “dangerous” for RFA (e.g., near the gallbladder or bowel loops). TACE should be preferred for treating lesions > 3 cm because its effectiveness appears to be better in well-vascularized tumors with large feeding arteries; selective and superselective TACE should be preferred, and the possible advantage of DEBs-TACE over lipiodol-TACE should be investigated in future studies. Multimodal treatment strategies, including sequentially applied TACE and RFA, appear to be promising, although the role of alternative treatments such as PLA, MWA, TARE, and CRT needs to be investigated in a larger number of patients. Regardless, all ablation procedures should be better evaluated with caution in patients with decompensated liver function to avoid irreversible liver failure and severe complications precluding LT.

The response of HCC to neoadjuvant treatments should be evaluated using the mRECIST criteria[118]. The RECIST criteria[124] were amended to the mRECIST in 2008[125] in the setting of HCC based on the concept that the evaluation of the treatment response should consider the amount of necrosis when estimating the decreased tumor load, not only the reduction in tumor size. However, it should also be considered that computed tomography and magnetic resonance imaging, which are currently used to assess the results of the ablation bridging procedures, tend to overestimate treatment effectiveness. In several studies, the concordance in the diagnosis of complete necrosis between the last imaging evaluation before LT and the pathological assessment at explant analysis of the target lesions has been reported to range between 50% and 83%; this is primarily due to the persistence of microscopic avascular neoplastic foci that are primarily located peripherally and cannot be detected by contrast-enhanced imaging techniques[74,75,77,85,106,115,126,127].

Although no solid conclusions may be drawn due to the absence of prospective comparative studies, it appears reasonable to state that bridging treatments decrease the dropout rate from the waiting list of T2-HCC patients and could have a positive impact on post-LT HCC recurrence and overall survival, at least in patients with complete or subtotal necrosis of the targeted lesions and a longer waiting period[29,105]. Furthermore, the response to pre-LT treatments may represent a surrogate marker of tumor biology and should be considered in the selection and prioritization of candidates for LT. That is, the transplant priority of T2-HCC candidates could be reduced after successful bridging therapy and a 3-6 mo period of observation confirming inactive neoplastic disease, and patients showing stable or progressive disease after treatment could then be prioritized. However, if an advantage is given on the waiting list to non-responding patients, a worsening in the outcome of LT in terms of overall survival, primarily due to an increased incidence of HCC recurrence after transplantation, should be considered. Whether this risk is acceptable is a matter of debate, and this issue should be further addressed in future studies[14,29,128].

HCC downstaging using exclusively TACE or multimodal sequential therapies to meet the conventional criteria for LT among carefully selected patients yields promising results in terms of overall and disease-free survival. In particular, some recent papers have demonstrated that patients successfully downstaged within the MC or the UCSF criteria can achieve a 5-year survival rate comparable to that of patients meeting the abovementioned criteria without requiring downstaging[123]. A follow-up period of 3 mo demonstrating stable disease after successful downstaging is suggested before inclusion on the waiting list for transplantation.

Footnotes

P- Reviewer: Cheung R, Kakizaki S, Tsuchiya A S- Editor: Cui XM L- Editor: A E- Editor: Liu XM

References
1.  Mazzaferro V, Regalia E, Doci R, Andreola S, Pulvirenti A, Bozzetti F, Montalto F, Ammatuna M, Morabito A, Gennari L. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996;334:693-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5110]  [Cited by in F6Publishing: 4991]  [Article Influence: 178.3]  [Reference Citation Analysis (0)]
2.  Llovet JM, Fuster J, Bruix J. Intention-to-treat analysis of surgical treatment for early hepatocellular carcinoma: resection versus transplantation. Hepatology. 1999;30:1434-1440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1331]  [Cited by in F6Publishing: 1393]  [Article Influence: 55.7]  [Reference Citation Analysis (0)]
3.  Yao FY, Bass NM, Nikolai B, Davern TJ, Kerlan R, Wu V, Ascher NL, Roberts JP. Liver transplantation for hepatocellular carcinoma: analysis of survival according to the intention-to-treat principle and dropout from the waiting list. Liver Transpl. 2002;8:873-883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in F6Publishing: 326]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
4.  Cillo U, Vitale A, Grigoletto F, Gringeri E, D’Amico F, Valmasoni M, Brolese A, Zanus G, Srsen N, Carraro A. Intention-to-treat analysis of liver transplantation in selected, aggressively treated HCC patients exceeding the Milan criteria. Am J Transplant. 2007;7:972-981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
5.  Kadry Z, Schaefer EW, Uemura T, Shah AR, Schreibman I, Riley TR. Impact of geographic disparity on liver allocation for hepatocellular cancer in the United States. J Hepatol. 2012;56:618-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
6.  Lai Q, Avolio AW, Lerut J, Singh G, Chan SC, Berloco PB, Tisone G, Agnes S, Chok KS, Sharr W. Recurrence of hepatocellular cancer after liver transplantation: the role of primary resection and salvage transplantation in East and West. J Hepatol. 2012;57:974-979.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 49]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
7.  Toso C, Dupuis-Lozeron E, Majno P, Berney T, Kneteman NM, Perneger T, Morel P, Mentha G, Combescure C. A model for dropout assessment of candidates with or without hepatocellular carcinoma on a common liver transplant waiting list. Hepatology. 2012;56:149-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 84]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
8.  Vitale A, Morales RR, Zanus G, Farinati F, Burra P, Angeli P, Frigo AC, Del Poggio P, Rapaccini G, Di Nolfo MA. Barcelona Clinic Liver Cancer staging and transplant survival benefit for patients with hepatocellular carcinoma: a multicentre, cohort study. Lancet Oncol. 2011;12:654-662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 109]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
9.  Avolio AW, Cillo U, Salizzoni M, De Carlis L, Colledan M, Gerunda GE, Mazzaferro V, Tisone G, Romagnoli R, Caccamo L. Balancing donor and recipient risk factors in liver transplantation: the value of D-MELD with particular reference to HCV recipients. Am J Transplant. 2011;11:2724-2736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 74]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
10.  Vitale A, D’Amico F, Frigo AC, Grigoletto F, Brolese A, Zanus G, Neri D, Carraro A, D’Amico FE, Burra P. Response to therapy as a criterion for awarding priority to patients with hepatocellular carcinoma awaiting liver transplantation. Ann Surg Oncol. 2010;17:2290-2302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
11.  Avolio AW, Siciliano M, Barbarino R, Nure E, Annicchiarico BE, Gasbarrini A, Agnes S, Castagneto M. Donor risk index and organ patient index as predictors of graft survival after liver transplantation. Transplant Proc. 2008;40:1899-1902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
12.  Wiesner RH, Freeman RB, Mulligan DC. Liver transplantation for hepatocellular cancer: the impact of the MELD allocation policy. Gastroenterology. 2004;127:S261-S267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 221]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
13.  Roayaie K, Feng S. Allocation policy for hepatocellular carcinoma in the MELD era: room for improvement? Liver Transpl. 2007;13:S36-S43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
14.  Cucchetti A, Cescon M, Bigonzi E, Piscaglia F, Golfieri R, Ercolani G, Cristina Morelli M, Ravaioli M, Daniele Pinna A. Priority of candidates with hepatocellular carcinoma awaiting liver transplantation can be reduced after successful bridge therapy. Liver Transpl. 2011;17:1344-1354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 77]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
15.  Del Gaudio M, Ercolani G, Ravaioli M, Cescon M, Lauro A, Vivarelli M, Zanello M, Cucchetti A, Vetrone G, Tuci F. Liver transplantation for recurrent hepatocellular carcinoma on cirrhosis after liver resection: University of Bologna experience. Am J Transplant. 2008;8:1177-1185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 129]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
16.  Freeman RB, Edwards EB, Harper AM. Waiting list removal rates among patients with chronic and malignant liver diseases. Am J Transplant. 2006;6:1416-1421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 162]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
17.  Washburn K, Edwards E, Harper A, Freeman R. Hepatocellular carcinoma patients are advantaged in the current liver transplant allocation system. Am J Transplant. 2010;10:1643-1648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 181]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
18.  Pomfret EA, Washburn K, Wald C, Nalesnik MA, Douglas D, Russo M, Roberts J, Reich DJ, Schwartz ME, Mieles L. Report of a national conference on liver allocation in patients with hepatocellular carcinoma in the United States. Liver Transpl. 2010;16:262-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 316]  [Cited by in F6Publishing: 291]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
19.  Cillo U, Vitale A, Volk ML, Frigo AC, Grigoletto F, Brolese A, Zanus G, D’Amico F, Farinati F, Burra P. The survival benefit of liver transplantation in hepatocellular carcinoma patients. Dig Liver Dis. 2010;42:642-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
20.  Ravaioli M, Grazi GL, Dazzi A, Bertuzzo V, Ercolani G, Cescon M, Cucchetti A, Masetti M, Ramacciato G, Pinna AD. Survival benefit after liver transplantation: a single European center experience. Transplantation. 2009;88:826-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
21.  Avolio AW, Siciliano M, Barone M, Lai Q, Caracciolo GL, Barbarino R, Nicolotti N, Lirosi MC, Gasbarrini A, Agnes S. Model for end-stage liver disease dynamic stratification of survival benefit. Transplant Proc. 2012;44:1851-1856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
22.  Merani S, Majno P, Kneteman NM, Berney T, Morel P, Mentha G, Toso C. The impact of waiting list α-fetoprotein changes on the outcome of liver transplant for hepatocellular carcinoma. J Hepatol. 2011;55:814-819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 143]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
23.  Mailey B, Artinyan A, Khalili J, Denitz J, Sanchez-Luege N, Sun CL, Bhatia S, Nissen N, Colquhoun SD, Kim J. Evaluation of absolute serum α-fetoprotein levels in liver transplant for hepatocellular cancer. Arch Surg. 2011;146:26-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
24.  De Giorgio M, Vezzoli S, Cohen E, Armellini E, Lucà MG, Verga G, Pinelli D, Nani R, Valsecchi MG, Antolini L. Prediction of progression-free survival in patients presenting with hepatocellular carcinoma within the Milan criteria. Liver Transpl. 2010;16:503-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
25.  Lai Q, Avolio AW, Graziadei I, Otto G, Rossi M, Tisone G, Goffette P, Vogel W, Pitton MB, Lerut J. α-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;19:1108-1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 152]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
26.  Knight M, Barber K, Gimson A, Collett D, Neuberger J. Implications of changing the minimal survival benefit in liver transplantation. Liver Transpl. 2012;18:549-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
27.  Merion RM, Schaubel DE, Dykstra DM, Freeman RB, Port FK, Wolfe RA. The survival benefit of liver transplantation. Am J Transplant. 2005;5:307-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 626]  [Cited by in F6Publishing: 575]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
28.  Schaubel DE, Sima CS, Goodrich NP, Feng S, Merion RM. The survival benefit of deceased donor liver transplantation as a function of candidate disease severity and donor quality. Am J Transplant. 2008;8:419-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 241]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
29.  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;58:609-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 108]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
30.  Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet. 2003;362:1907-1917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3241]  [Cited by in F6Publishing: 3212]  [Article Influence: 153.0]  [Reference Citation Analysis (0)]
31.  Silva MF, Sherman M. Criteria for liver transplantation for HCC: what should the limits be? J Hepatol. 2011;55:1137-1147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
32.  Yao FY, Ferrell L, Bass NM, Watson JJ, Bacchetti P, Venook A, Ascher NL, Roberts JP. Liver transplantation for hepatocellular carcinoma: expansion of the tumor size limits does not adversely impact survival. Hepatology. 2001;33:1394-1403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1594]  [Cited by in F6Publishing: 1583]  [Article Influence: 68.8]  [Reference Citation Analysis (0)]
33.  Mazzaferro V, Llovet JM, Miceli R, Bhoori S, Schiavo M, Mariani L, Camerini T, Roayaie S, Schwartz ME, Grazi GL. Predicting survival after liver transplantation in patients with hepatocellular carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet Oncol. 2009;10:35-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1267]  [Cited by in F6Publishing: 1421]  [Article Influence: 88.8]  [Reference Citation Analysis (1)]
34.  Toso C, Mentha G, Kneteman NM, Majno P. The place of downstaging for hepatocellular carcinoma. J Hepatol. 2010;52:930-936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 106]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
35.  Sala M, Fuster J, Llovet JM, Navasa M, Solé M, Varela M, Pons F, Rimola A, García-Valdecasas JC, Brú C. High pathological risk of recurrence after surgical resection for hepatocellular carcinoma: an indication for salvage liver transplantation. Liver Transpl. 2004;10:1294-1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 243]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
36.  Earl TM, Chapman WC. Hepatocellular Carcinoma: resection versus transplantation. Semin Liver Dis. 2013;33:282-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
37.  Lei JY, Yan LN, Wang WT. Transplantation vs resection for hepatocellular carcinoma with compensated liver function after downstaging therapy. World J Gastroenterol. 2013;19:4400-4408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 25]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
38.  Lee KK, Kim DG, Moon IS, Lee MD, Park JH. Liver transplantation versus liver resection for the treatment of hepatocellular carcinoma. J Surg Oncol. 2010;101:47-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 108]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
39.  Cillo U, Vitale A, Dupuis D, Corso S, Neri D, D’Amico F, Gringeri E, Farinati F, Vincenzi V, Zanus G. Laparoscopic ablation of hepatocellular carcinoma in cirrhotic patients unsuitable for liver resection or percutaneous treatment: a cohort study. PLoS One. 2013;8:e57249.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Majno PE, Sarasin FP, Mentha G, Hadengue A. Primary liver resection and salvage transplantation or primary liver transplantation in patients with single, small hepatocellular carcinoma and preserved liver function: an outcome-oriented decision analysis. Hepatology. 2000;31:899-906.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Barone M, Avolio AW, Di Leo A, Burra P, Francavilla A. ABO blood group-related waiting list disparities in liver transplant candidates: effect of the MELD adoption. Transplantation. 2008;85:844-849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
42.  Lei J, Yan L, Wang W. Comparison of the outcomes of patients who underwent deceased-donor or living-donor liver transplantation after successful downstaging therapy. Eur J Gastroenterol Hepatol. 2013;25:1340-1346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
43.  Grant RC, Sandhu L, Dixon PR, Greig PD, Grant DR, McGilvray ID. Living vs. deceased donor liver transplantation for hepatocellular carcinoma: a systematic review and meta-analysis. Clin Transplant. 2013;27:140-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 70]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
44.  Avolio AW, Halldorson JB, Burra P, Dutkowski P, Agnes S, Clavien PA. Balancing utility and need by means of donor-to-recipient matching: a challenging problem. Am J Transplant. 2013;13:522-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
45.  Adam R, Azoulay D, Castaing D, Eshkenazy R, Pascal G, Hashizume K, Samuel D, Bismuth H. Liver resection as a bridge to transplantation for hepatocellular carcinoma on cirrhosis: a reasonable strategy? Ann Surg. 2003;238:508-518; discussion 518-519.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 254]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
46.  Belghiti J, Cortes A, Abdalla EK, Régimbeau JM, Prakash K, Durand F, Sommacale D, Dondero F, Lesurtel M, Sauvanet A. Resection prior to liver transplantation for hepatocellular carcinoma. Ann Surg. 2003;238:885-892; discussion 892-893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 342]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
47.  Facciuto ME, Koneru B, Rocca JP, Wolf DC, Kim-Schluger L, Visintainer P, Klein KM, Chun H, Marvin M, Rozenblit G. Surgical treatment of hepatocellular carcinoma beyond Milan criteria. Results of liver resection, salvage transplantation, and primary liver transplantation. Ann Surg Oncol. 2008;15:1383-1391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 66]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
48.  Liu F, Wei Y, Wang W, Chen K, Yan L, Wen T, Zhao J, Xu M, Li B. Salvage liver transplantation for recurrent hepatocellular carcinoma within UCSF criteria after liver resection. PLoS One. 2012;7:e48932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
49.  Abecassis MM, Fisher RA, Olthoff KM, Freise CE, Rodrigo DR, Samstein B, Kam I, Merion RM. Complications of living donor hepatic lobectomy--a comprehensive report. Am J Transplant. 2012;12:1208-1217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 234]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
50.  Hwang S, Lee SG, Moon DB, Ahn CS, Kim KH, Lee YJ, Ha TY, Song GW. Salvage living donor liver transplantation after prior liver resection for hepatocellular carcinoma. Liver Transpl. 2007;13:741-746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 101]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
51.  Kaido T, Mori A, Ogura Y, Hata K, Yoshizawa A, Iida T, Yagi S, Uemoto S. Living donor liver transplantation for recurrent hepatocellular carcinoma after liver resection. Surgery. 2012;151:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
52.  Moon JI, Kwon CH, Joh JW, Choi GS, Jung GO, Kim JM, Shin M, Choi SJ, Kim SJ, Lee SK. Primary versus salvage living donor liver transplantation for patients with hepatocellular carcinoma: impact of microvascular invasion on survival. Transplant Proc. 2012;44:487-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
53.  Poon RT, Fan ST, Lo CM, Liu CL, Wong J. Long-term survival and pattern of recurrence after resection of small hepatocellular carcinoma in patients with preserved liver function: implications for a strategy of salvage transplantation. Ann Surg. 2002;235:373-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 647]  [Cited by in F6Publishing: 664]  [Article Influence: 30.2]  [Reference Citation Analysis (0)]
54.  Llovet JM, Brú C, Bruix J. Prognosis of hepatocellular carcinoma: the BCLC staging classification. Semin Liver Dis. 1999;19:329-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2645]  [Cited by in F6Publishing: 2715]  [Article Influence: 108.6]  [Reference Citation Analysis (0)]
55.  Llovet JM, Bruix J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology. 2003;37:429-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2207]  [Cited by in F6Publishing: 2176]  [Article Influence: 103.6]  [Reference Citation Analysis (0)]
56.  Marelli L, Stigliano R, Triantos C, Senzolo M, Cholongitas E, Davies N, Tibballs J, Meyer T, Patch DW, Burroughs AK. Transarterial therapy for hepatocellular carcinoma: which technique is more effective? A systematic review of cohort and randomized studies. Cardiovasc Intervent Radiol. 2007;30:6-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 590]  [Article Influence: 34.7]  [Reference Citation Analysis (0)]
57.  Varela M, Real MI, Burrel M, Forner A, Sala M, Brunet M, Ayuso C, Castells L, Montañá X, Llovet JM. Chemoembolization of hepatocellular carcinoma with drug eluting beads: efficacy and doxorubicin pharmacokinetics. J Hepatol. 2007;46:474-481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 694]  [Cited by in F6Publishing: 749]  [Article Influence: 44.1]  [Reference Citation Analysis (1)]
58.  Lammer J, Malagari K, Vogl T, Pilleul F, Denys A, Watkinson A, Pitton M, Sergent G, Pfammatter T, Terraz S. Prospective randomized study of doxorubicin-eluting-bead embolization in the treatment of hepatocellular carcinoma: results of the PRECISION V study. Cardiovasc Intervent Radiol. 2010;33:41-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1063]  [Cited by in F6Publishing: 1115]  [Article Influence: 74.3]  [Reference Citation Analysis (0)]
59.  Majno PE, Adam R, Bismuth H, Castaing D, Ariche A, Krissat J, Perrin H, Azoulay D. Influence of preoperative transarterial lipiodol chemoembolization on resection and transplantation for hepatocellular carcinoma in patients with cirrhosis. Ann Surg. 1997;226:688-701; discussion 701-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 383]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
60.  Graziadei IW, Sandmueller H, Waldenberger P, Koenigsrainer A, Nachbaur K, Jaschke W, Margreiter R, Vogel W. Chemoembolization followed by liver transplantation for hepatocellular carcinoma impedes tumor progression while on the waiting list and leads to excellent outcome. Liver Transpl. 2003;9:557-563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 347]  [Cited by in F6Publishing: 326]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
61.  Hayashi PH, Ludkowski M, Forman LM, Osgood M, Johnson S, Kugelmas M, Trotter JF, Bak T, Wachs M, Kam I. Hepatic artery chemoembolization for hepatocellular carcinoma in patients listed for liver transplantation. Am J Transplant. 2004;4:782-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 66]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
62.  Decaens T, Roudot-Thoraval F, Bresson-Hadni S, Meyer C, Gugenheim J, Durand F, Bernard PH, Boillot O, Boudjema K, Calmus Y. Impact of pretransplantation transarterial chemoembolization on survival and recurrence after liver transplantation for hepatocellular carcinoma. Liver Transpl. 2005;11:767-775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 159]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
63.  Millonig G, Graziadei IW, Freund MC, Jaschke W, Stadlmann S, Ladurner R, Margreiter R, Vogel W. Response to preoperative chemoembolization correlates with outcome after liver transplantation in patients with hepatocellular carcinoma. Liver Transpl. 2007;13:272-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 198]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
64.  Alba E, Valls C, Dominguez J, Martinez L, Escalante E, Lladó L, Serrano T. Transcatheter arterial chemoembolization in patients with hepatocellular carcinoma on the waiting list for orthotopic liver transplantation. AJR Am J Roentgenol. 2008;190:1341-1348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
65.  Golfieri R, Cappelli A, Cucchetti A, Piscaglia F, Carpenzano M, Peri E, Ravaioli M, D’Errico-Grigioni A, Pinna AD, Bolondi L. Efficacy of selective transarterial chemoembolization in inducing tumor necrosis in small (& lt; 5 cm) hepatocellular carcinomas. Hepatology. 2011;53:1580-1589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
66.  Kwan SW, Fidelman N, Ma E, Kerlan RK, Yao FY. Imaging predictors of the response to transarterial chemoembolization in patients with hepatocellular carcinoma: a radiological-pathological correlation. Liver Transpl. 2012;18:727-736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 88]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
67.  Tsochatzis E, Garcovich M, Marelli L, Papastergiou V, Fatourou E, Rodriguez-Peralvarez ML, Germani G, Davies N, Yu D, Luong TV. Transarterial embolization as neo-adjuvant therapy pretransplantation in patients with hepatocellular carcinoma. Liver Int. 2013;33:944-949.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
68.  Nicolini A, Martinetti L, Crespi S, Maggioni M, Sangiovanni A. Transarterial chemoembolization with epirubicin-eluting beads versus transarterial embolization before liver transplantation for hepatocellular carcinoma. J Vasc Interv Radiol. 2010;21:327-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 109]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
69.  Majno P, Giostra E, Mentha G. Management of hepatocellular carcinoma on the waiting list before liver transplantation: time for controlled trials? Liver Transpl. 2007;13:S27-S35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
70.  Richard HM, Silberzweig JE, Mitty HA, Lou WY, Ahn J, Cooper JM. Hepatic arterial complications in liver transplant recipients treated with pretransplantation chemoembolization for hepatocellular carcinoma. Radiology. 2000;214:775-779.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Pérez Saborido B, Meneu JC, Moreno E, García I, Moreno A, Fundora Y. Is transarterial chemoembolization necessary before liver transplantation for hepatocellular carcinoma? Am J Surg. 2005;190:383-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
72.  Shibata T, Shibata T, Maetani Y, Isoda H, Hiraoka M. Radiofrequency ablation for small hepatocellular carcinoma: prospective comparison of internally cooled electrode and expandable electrode. Radiology. 2006;238:346-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 99]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
73.  Mazzaferro V, Battiston C, Perrone S, Pulvirenti A, Regalia E, Romito R, Sarli D, Schiavo M, Garbagnati F, Marchianò A. Radiofrequency ablation of small hepatocellular carcinoma in cirrhotic patients awaiting liver transplantation: a prospective study. Ann Surg. 2004;240:900-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 369]  [Cited by in F6Publishing: 406]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
74.  Lu DS, Yu NC, Raman SS, Lassman C, Tong MJ, Britten C, Durazo F, Saab S, Han S, Finn R. Percutaneous radiofrequency ablation of hepatocellular carcinoma as a bridge to liver transplantation. Hepatology. 2005;41:1130-1137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 256]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
75.  Pompili M, Mirante VG, Rondinara G, Fassati LR, Piscaglia F, Agnes S, Covino M, Ravaioli M, Fagiuoli S, Gasbarrini G. 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;11:1117-1126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 166]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
76.  Brillet PY, Paradis V, Brancatelli G, Rangheard AS, Consigny Y, Plessier A, Durand F, Belghiti J, Sommacale D, Vilgrain V. Percutaneous radiofrequency ablation for hepatocellular carcinoma before liver transplantation: a prospective study with histopathologic comparison. AJR Am J Roentgenol. 2006;186:S296-S305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 61]  [Article Influence: 3.6]  [Reference Citation Analysis (1)]
77.  Rodríguez-Sanjuán JC, González F, Juanco C, Herrera LA, López-Bautista M, González-Noriega M, García-Somacarrera E, Figols J, Gómez-Fleitas M, Silván M. Radiological and pathological assessment of hepatocellular carcinoma response to radiofrequency. A study on removed liver after transplantation. World J Surg. 2008;32:1489-1494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 25]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
78.  DuBay DA, Sandroussi C, Kachura JR, Ho CS, Beecroft JR, Vollmer CM, Ghanekar A, Guba M, Cattral MS, McGilvray ID. Radiofrequency ablation of hepatocellular carcinoma as a bridge to liver transplantation. HPB (Oxford). 2011;13:24-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
79.  Porrett PM, Peterman H, Rosen M, Sonnad S, Soulen M, Markmann JF, Shaked A, Furth E, Reddy KR, Olthoff K. Lack of benefit of pre-transplant locoregional hepatic therapy for hepatocellular cancer in the current MELD era. Liver Transpl. 2006;12:665-673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 110]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
80.  Lopez KT, Kuwada SK, Wong LL. Consequences of needle tract seeding of hepatocellular cancer after liver transplant. Clin Transplant. 2013;27:E400-E406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
81.  Castroagudín JF, Delgado M, Villanueva A, Bustamante M, Martínez J, Otero E, Tomé S, Martínez SM, Segade FR, Conde R. Safety of percutaneous ethanol injection as neoadjuvant therapy for hepatocellular carcinoma in waiting list liver transplant candidates. Transplant Proc. 2005;37:3871-3873.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
82.  Branco F, Brú C, Vilana R, Bianchi L, Alves de Mattos A. Percutaneous ethanol injection before liver transplantation in the hepatocellular carcinoma. Ann Hepatol. 2009;8:220-227.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Pacella CM, Bizzarri G, Francica G, Forlini G, Petrolati A, Valle D, Anelli V, Bianchini A, Nuntis SD, Pacella S. Analysis of factors predicting survival in patients with hepatocellular carcinoma treated with percutaneous laser ablation. J Hepatol. 2006;44:902-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
84.  Gough-Palmer AL, Gedroyc WM. Laser ablation of hepatocellular carcinoma--a review. World J Gastroenterol. 2008;14:7170-7174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 65]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
85.  Pompili M, Pacella CM, Francica G, Angelico M, Tisone G, Craboledda P, Nicolardi E, Rapaccini GL, Gasbarrini G. Percutaneous laser ablation of hepatocellular carcinoma in patients with liver cirrhosis awaiting liver transplantation. Eur J Radiol. 2010;74:e6-e11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
86.  Francica G, Petrolati A, Di Stasio E, Pacella S, Stasi R, Pacella CM. Effectiveness, safety, and local progression after percutaneous laser ablation for hepatocellular carcinoma nodules up to 4 cm are not affected by tumor location. AJR Am J Roentgenol. 2012;199:1393-1401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
87.  Lu MD, Xu HX, Xie XY, Yin XY, Chen JW, Kuang M, Xu ZF, Liu GJ, Zheng YL. Percutaneous microwave and radiofrequency ablation for hepatocellular carcinoma: a retrospective comparative study. J Gastroenterol. 2005;40:1054-1060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 166]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
88.  Boutros C, Somasundar P, Garrean S, Saied A, Espat NJ. Microwave coagulation therapy for hepatic tumors: review of the literature and critical analysis. Surg Oncol. 2010;19:e22-e32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 108]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
89.  Zanus G, Boetto R, Gringeri E, Vitale A, D’Amico F, Carraro A, Bassi D, Bonsignore P, Noaro G, Mescoli C. Microwave thermal ablation for hepatocarcinoma: six liver transplantation cases. Transplant Proc. 2011;43:1091-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
90.  Riaz A, Kulik L, Lewandowski RJ, Ryu RK, Giakoumis Spear G, Mulcahy MF, Abecassis M, Baker T, Gates V, Nayar R. Radiologic-pathologic correlation of hepatocellular carcinoma treated with internal radiation using yttrium-90 microspheres. Hepatology. 2009;49:1185-1193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 223]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
91.  Sandroussi C, Dawson LA, Lee M, Guindi M, Fischer S, Ghanekar A, Cattral MS, McGilvray ID, Levy GA, Renner E. Radiotherapy as a bridge to liver transplantation for hepatocellular carcinoma. Transpl Int. 2010;23:299-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 81]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
92.  Rossi S, Garbagnati F, Lencioni R, Allgaier HP, Marchianò A, Fornari F, Quaretti P, Tolla GD, Ambrosi C, Mazzaferro V. Percutaneous radio-frequency thermal ablation of nonresectable hepatocellular carcinoma after occlusion of tumor blood supply. Radiology. 2000;217:119-126.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Veltri A, Moretto P, Doriguzzi A, Pagano E, Carrara G, Gandini G. Radiofrequency thermal ablation (RFA) after transarterial chemoembolization (TACE) as a combined therapy for unresectable non-early hepatocellular carcinoma (HCC). Eur Radiol. 2006;16:661-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 157]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
94.  Ashoori N, Paprottka P, Trumm C, Bamberg F, Kolligs FT, Rentsch M, Reiser MF, Jakobs TF. Multimodality treatment with conventional transcatheter arterial chemoembolization and radiofrequency ablation for unresectable hepatocellular carcinoma. Digestion. 2012;85:18-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
95.  Iezzi R, Cesario V, Siciliani L, Campanale M, De Gaetano AM, Siciliano M, Agnes S, Giuliante F, Grieco A, Pompili M. Single-step multimodal locoregional treatment for unresectable hepatocellular carcinoma: balloon-occluded percutaneous radiofrequency thermal ablation (BO-RFA) plus transcatheter arterial chemoembolization (TACE). Radiol Med. 2013;118:555-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
96.  Ashoori N, Bamberg F, Paprottka P, Rentsch M, Kolligs FT, Siegert S, Peporte A, Al-Tubaikh JA, D’Anastasi M, Hoffmann RT. Multimodality treatment for early-stage hepatocellular carcinoma: a bridging therapy for liver transplantation. Digestion. 2012;86:338-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
97.  Lesurtel M, Müllhaupt B, Pestalozzi BC, Pfammatter T, Clavien PA. Transarterial chemoembolization as a bridge to liver transplantation for hepatocellular carcinoma: an evidence-based analysis. Am J Transplant. 2006;6:2644-2650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 138]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
98.  Maddala YK, Stadheim L, Andrews JC, Burgart LJ, Rosen CB, Kremers WK, Gores G. Drop-out rates of patients with hepatocellular cancer listed for liver transplantation: outcome with chemoembolization. Liver Transpl. 2004;10:449-455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 177]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
99.  Fontana RJ, Hamidullah H, Nghiem H, Greenson JK, Hussain H, Marrero J, Rudich S, McClure LA, Arenas J. Percutaneous radiofrequency thermal ablation of hepatocellular carcinoma: a safe and effective bridge to liver transplantation. Liver Transpl. 2002;8:1165-1174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 139]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
100.  Ciccarelli O, Lai Q, Goffette P, Finet P, De Reyck C, Roggen F, Sempoux C, Doffagne E, Reding R, Lerut J. Liver transplantation for hepatocellular cancer: UCL experience in 137 adult cirrhotic patients. α-foetoprotein level and locoregional treatment as refined selection criteria. Transpl Int. 2012;25:867-875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
101.  Kornberg A, Witt U, Matevossian E, Küpper B, Assfalg V, Drzezga A, Hüser N, Wildgruber M, Friess H. Extended postinterventional tumor necrosis-implication for outcome in liver transplant patients with advanced HCC. PLoS One. 2013;8:e53960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
102.  Bharat A, Brown DB, Crippin JS, Gould JE, Lowell JA, Shenoy S, Desai NM, Chapman WC. Pre-liver transplantation locoregional adjuvant therapy for hepatocellular carcinoma as a strategy to improve longterm survival. J Am Coll Surg. 2006;203:411-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 83]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
103.  Lao OB, Weissman J, Perkins JD. Pre-transplant therapy for hepatocellular carcinoma is associated with a lower recurrence after liver transplantation. Clin Transplant. 2009;23:874-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
104.  Heckman JT, Devera MB, Marsh JW, Fontes P, Amesur NB, Holloway SE, Nalesnik M, Geller DA, Steel JL, Gamblin TC. Bridging locoregional therapy for hepatocellular carcinoma prior to liver transplantation. Ann Surg Oncol. 2008;15:3169-3177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 111]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
105.  Freeman RB, Steffick DE, Guidinger MK, Farmer DG, Berg CL, Merion RM. Liver and intestine transplantation in the United States, 1997-2006. Am J Transplant. 2008;8:958-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 207]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
106.  Yao FY, Kerlan RK, Hirose R, Davern TJ, Bass NM, Feng S, Peters M, Terrault N, Freise CE, Ascher NL. Excellent outcome following down-staging of hepatocellular carcinoma prior to liver transplantation: an intention-to-treat analysis. Hepatology. 2008;48:819-827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 386]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
107.  De Luna W, Sze DY, Ahmed A, Ha BY, Ayoub W, Keeffe EB, Cooper A, Esquivel C, Nguyen MH. Transarterial chemoinfusion for hepatocellular carcinoma as downstaging therapy and a bridge toward liver transplantation. Am J Transplant. 2009;9:1158-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 104]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
108.  Chapman WC, Majella Doyle MB, Stuart JE, Vachharajani N, Crippin JS, Anderson CD, Lowell JA, Shenoy S, Darcy MD, Brown DB. Outcomes of neoadjuvant transarterial chemoembolization to downstage hepatocellular carcinoma before liver transplantation. Ann Surg. 2008;248:617-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 225]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
109.  Lewandowski RJ, Kulik LM, Riaz A, Senthilnathan S, Mulcahy MF, Ryu RK, Ibrahim SM, Sato KT, Baker T, Miller FH. A comparative analysis of transarterial downstaging for hepatocellular carcinoma: chemoembolization versus radioembolization. Am J Transplant. 2009;9:1920-1928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 408]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
110.  Jang JW, You CR, Kim CW, Bae SH, Yoon SK, Yoo YK, Kim DG, Choi JY. Benefit of downsizing hepatocellular carcinoma in a liver transplant population. Aliment Pharmacol Ther. 2010;31:415-423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 47]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
111.  Barakat O, Wood RP, Ozaki CF, Ankoma-Sey V, Galati J, Skolkin M, Toombs B, Round M, Moore W, Mieles L. Morphological features of advanced hepatocellular carcinoma as a predictor of downstaging and liver transplantation: an intention-to-treat analysis. Liver Transpl. 2010;16:289-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 73]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
112.  Bargellini I, Vignali C, Cioni R, Petruzzi P, Cicorelli A, Campani D, De Simone P, Filipponi F, Bartolozzi C. Hepatocellular carcinoma: CT for tumor response after transarterial chemoembolization in patients exceeding Milan criteria--selection parameter for liver transplantation. Radiology. 2010;255:289-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
113.  Bova V, Miraglia R, Maruzzelli L, Vizzini GB, Luca A. Predictive factors of downstaging of hepatocellular carcinoma beyond the Milan criteria treated with intra-arterial therapies. Cardiovasc Intervent Radiol. 2013;36:433-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
114.  Lei J, Wang W, Yan L. Downstaging advanced hepatocellular carcinoma to the milan criteria may provide a comparable outcome to conventional Milan criteria. J Gastrointest Surg. 2013;17:1440-1446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
115.  Ravaioli M, Grazi GL, Piscaglia F, Trevisani F, Cescon M, Ercolani G, Vivarelli M, Golfieri R, D’Errico Grigioni A, Panzini I. Liver transplantation for hepatocellular carcinoma: results of down-staging in patients initially outside the Milan selection criteria. Am J Transplant. 2008;8:2547-2557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 271]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
116.  Otto G, Herber S, Heise M, Lohse AW, Mönch C, Bittinger F, Hoppe-Lotichius M, Schuchmann M, Victor A, Pitton M. Response to transarterial chemoembolization as a biological selection criterion for liver transplantation in hepatocellular carcinoma. Liver Transpl. 2006;12:1260-1267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 304]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
117.  DuBay D, Sandroussi C, Sandhu L, Cleary S, Guba M, Cattral MS, McGilvray I, Ghanekar A, Selzner M, Greig PD. Liver transplantation for advanced hepatocellular carcinoma using poor tumor differentiation on biopsy as an exclusion criterion. Ann Surg. 2011;253:166-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 210]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
118.  Lencioni R, Llovet JM. Modified RECIST (mRECIST) assessment for hepatocellular carcinoma. Semin Liver Dis. 2010;30:52-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2583]  [Cited by in F6Publishing: 2894]  [Article Influence: 206.7]  [Reference Citation Analysis (36)]
119.  Gordon-Weeks AN, Snaith A, Petrinic T, Friend PJ, Burls A, Silva MA. Systematic review of outcome of downstaging hepatocellular cancer before liver transplantation in patients outside the Milan criteria. Br J Surg. 2011;98:1201-1208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
120.  Belghiti J, Carr BI, Greig PD, Lencioni R, Poon RT. Treatment before liver transplantation for HCC. Ann Surg Oncol. 2008;15:993-1000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 101]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
121.  Bruix J, Sherman M. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53:1020-1022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5972]  [Cited by in F6Publishing: 6326]  [Article Influence: 486.6]  [Reference Citation Analysis (1)]
122.  European Association For The Study Of The Liver; European Organisation For Research And Treatment Of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908-943.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  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:e11-e22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 673]  [Cited by in F6Publishing: 728]  [Article Influence: 56.0]  [Reference Citation Analysis (1)]
124.  Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92:205-216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12751]  [Cited by in F6Publishing: 12879]  [Article Influence: 536.6]  [Reference Citation Analysis (0)]
125.  Llovet JM, Di Bisceglie AM, Bruix J, Kramer BS, Lencioni R, Zhu AX, Sherman M, Schwartz M, Lotze M, Talwalkar J. Design and endpoints of clinical trials in hepatocellular carcinoma. J Natl Cancer Inst. 2008;100:698-711.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1232]  [Cited by in F6Publishing: 1298]  [Article Influence: 81.1]  [Reference Citation Analysis (0)]
126.  Kim YS, Rhim H, Lim HK, Park CK, Lee WJ, Do YS, Cho JW. Completeness of treatment in hepatocellular carcinomas treated with image-guided tumor therapies: Evaluation of positive predictive value of contrast-enhanced CT with histopathologic correlation in the explanted liver specimen. J Comput Assist Tomogr. 2006;30:578-582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
127.  Bargellini I, Bozzi E, Campani D, Carrai P, De Simone P, Pollina L, Cioni R, Filipponi F, Bartolozzi C. Modified RECIST to assess tumor response after transarterial chemoembolization of hepatocellular carcinoma: CT-pathologic correlation in 178 liver explants. Eur J Radiol. 2013;82:e212-e218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
128.  Roberts JP, Venook A, Kerlan R, Yao F. Hepatocellular carcinoma: Ablate and wait versus rapid transplantation. Liver Transpl. 2010;16:925-929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 108]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]