Minireviews Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. May 18, 2015; 7(8): 1142-1148
Published online May 18, 2015. doi: 10.4254/wjh.v7.i8.1142
Management of recurrent hepatocellular carcinoma after liver transplant
Kenneth SH Chok, Department of Surgery, The University of Hong Kong, Hong Kong, China
Author contributions: Chok KSH solely contributed to this paper.
Conflict-of-interest: The author has no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Kenneth SH Chok, MBBS, Department of Surgery, The University of Hong Kong, 102 Pok Fu Lam Road, Hong Kong, China. kennethchok@gmail.com
Telephone: +852-2255-3025 Fax: +852-2816-5284
Received: September 5, 2014
Peer-review started: September 6, 2014
First decision: January 8, 2015
Revised: January 27, 2015
Accepted: February 10, 2015
Article in press: February 12, 2015
Published online: May 18, 2015

Abstract

Hepatocellular carcinoma (HCC) is the leading cause of deaths in patients with hepatitis B or C, and its incidence has increased considerably over the past decade and is still on the rise. Liver transplantation (LT) provides the best chance of cure for patients with HCC and liver cirrhosis. With the implementation of the MELD exception system for patients with HCC waitlisted for LT, the number of recipients of LT is increasing, so is the number of patients who have recurrence of HCC after LT. Treatments for intrahepatic recurrence after transplantation and after other kinds of surgery are more or less the same, but long-term cure of posttransplant recurrence is rarely seen as it is a “systemic” disease. Nonetheless, surgical resection has been shown to be effective in prolonging patient survival despite the technical difficulty in resecting graft livers. Besides surgical resection, different kinds of treatment are also in use, including transarterial chemoembolization, radiofrequency ablation, high-intensity focused ultrasound ablation, and stereotactic body radiation therapy. Targeted therapy and modulation of immunosuppressants are also adopted to treat the deadly disease.

Key Words: Hepatocellular carcinoma, Recurrence, Transarterial chemoembolization, Liver transplantation, Targeted therapy, Resection, Radiofrequency ablation, Transarterial radioembolization, Immunosuppression, Stereotactic body radiation therapy

Core tip: The management of recurrent hepatocellular carcinoma (HCC) after liver transplantation (LT) seems to be a losing battle. Nonetheless, tremendous efforts have been made to combat this deadly disease. Intrahepatic recurrence may be treated by resection, which has some survival benefits as shown by small clinical trials. Other kinds of therapy including high-intensity focused ultrasound (HIFU) ablation, radiofrequency ablation (RFA) and transarterial chemoembolization (TACE) are also in use. HIFU ablation has been shown to produce better results when compared with RFA and TACE. The efficacy of systemic and targeted therapies for multiple recurrences is under investigation. Early results have suggested that the combination of sorafenib with mammalian target of rapamycin inhibitors may be useful for treating recurrent HCC after LT.



INTRODUCTION

Hepatocellular carcinoma (HCC) is the fifth most common malignant tumor, the third leading cause of cancer-related deaths, and the first leading cause of deaths in patients with hepatitis B or C, and its incidence has increased considerably over the past decade and is still on the rise[1-3]. There are different modalities for treating HCC and underlying liver cirrhosis, but liver transplantation (LT) is the ultimate solution[4]. Various patient selection criteria for LT have been introduced with the hope that as many patients as possible can benefit from the treatment while patient survival is not compromised. Mazzaferro et al[5] introduced the Milan criteria (solitary tumor ≤ 5 cm, or ≤ 3 tumors with each measuring < 3 cm) on the basis of a retrospective study of 48 patients who received LT for HCC. In the study, a 75% overall survival and an 83% recurrence-free survival were achieved in LT recipients chosen according to the Milan criteria at 4 years after transplantation. A set of modestly expanded criteria was developed by the University of California, San Francisco (UCSF). Yao et al[6] showed that HCC patients selected for LT according to the UCSF criteria (solitary tumor ≤ 6.5 cm, or ≤ 3 nodules with the largest lesion ≤ 4.5 cm and a total tumor diameter ≤ 8 cm) had survival rates of 90% and 75.2% at 1 year and 5 years respectively. However, discrepancy between radiological results and pathological results of tumor characteristics is not uncommon. A 30%-50% discrepancy rate has been reported[6,7].

In Hong Kong, about 8% of the population are carriers of hepatitis B virus (HBV) and most of the cases of HCC are caused by HBV. A survey found that about 10.4% of male adults and 7.7% of female adults were positive of hepatitis B surface antigen (surveillance of viral hepatitis in Hong Kong - 2010 update report. Hong Kong SAR: Department of Health, 2011). On the other hand, the numbers of carriers of hepatitis C virus (HCV) are rising in Japan and the United States. In these places where hepatitis C is epidemic, there is a surge of HCV-related liver cirrhosis and HCC[8,9].

Even though HCC patients are selected for LT according to standard criteria, 10%-60% of them will have disease recurrence. Some of them will develop recurrence 2 years or even 5 years after transplantation[10]. With the adoption of the MELD exception system for HCC patients waitlisted for LT, more LTs are performed for HCC. Hong Kong adopted the system in 2009[11], and nowadays HCC accounts for one third of LTs in Hong Kong. As a corollary, the incidence of HCC recurrence after LT is on the increase in places where the system is adopted. Recurrence of HCC after LT is notoriously difficult to manage. Here is a review of the treatment options available for this challenging situation, trying to shed some light on its management.

RISK FACTORS FOR HCC RECURRENCE

Post-LT HCC recurrence occurs at a rate of 13%-27%[10,12]. It was reported that 5% of patients developed late (after 5 years) recurrence[10]. Most patient selection criteria for LT, including the Milan and the UCSF criteria, use tumor size and tumor number as surrogate markers. A meta-analysis by Sotiropoulos et al[13] identified a number of risk factors for poorer patient survival after LT, which were venous invasion, poor tumor cell differentiation, tumor size and stage beyond the Milan criteria, and a high pretransplant serum α-fetoprotein level. Since radiological results and pathological results of tumor characteristics may differ, some centers use pretransplant serum α-fetoprotein level and biopsy to determine tumor cell differentiation and use it as a biological surrogate marker in patient selection criteria[14,15]. However, preoperative biopsy may cause tumor seeding and bleeding. Saborido et al[16] reported that a significantly higher chance of HCC recurrence came with fine-needle aspiration biopsy before LT (31.8% vs 5.9%, P = 0.003). In Hong Kong, contrast computed tomography (CT)[17] is used for tumor staging. Sometimes positron emission tomography (PET) using both radiotracers of 11C-acetate and 18F-FDG is also employed. In a report, dual-tracer PET had an overall sensitivity of 96.8% and an overall specificity of 91.7%, which are significantly higher than those of contrast CT (41.9% and 33.0% respectively; P < 0.05 in both cases)[18]. It was found that sources of error for contrast CT were related to liver cirrhosis or previous treatment, and there was difficulty in differentiating cirrhotic nodules from HCCs (39%) and in the estimation of tumor size (14%). There was infrequent overstaging of vascular invasion (4.6%) or extrahepatic metastasis (4.6%). Dual-tracer PET and contrast CT had a 4.7% rate of false-negative results. PET using the radiotracer 18F-FDG seems effective in detecting 18F-FDG-avid lesions and thus can be used as an adjunct to detect microvascular invasion[19]. Nonetheless, such use is still at its infancy and more large-scale trials are needed for its validation.

Deceased-donor LT vs living-donor LT

Living-donor LT (LDLT) has the most significant impact in Asia, where the issue of organ shortage is most extreme. The availability of LDLT has provided the driving force for a drastic increase in cases of LT in recent years. The number of LDLTs performed in Asia each year has increased tremendously. In 2005, LDLT accounted for 90% of the 1497 LTs performed in Asia (excluding mainland China)[20]. In Hong Kong, about half of the LTs are LDLTs, and more than half are for HCC.

To justify LDLT for HCC, it should have a survival outcome comparable to that of deceased-donor LT (DDLT). Roayaie et al[21] reported a tendency for early tumor recurrence after LDLT (mean: 8.7 mo) when compared with DDLT (mean: 19.6 mo) in a cohort of 311 patients with histologically confirmed HCC after LT. Another multicenter LDLT cohort study (A2ALL) of 106 HCC patients reported a significantly higher 3-year tumor recurrence rate after LDLT (29%) compared with that after DDLT (0%)[22]. In Hong Kong, a retrospective study has been conducted to compare LDLT and DDLT in terms of treatment outcomes in 60 HCC patients[23]. Given the standard patient selection criteria based on radiological tumor size and number according to the UCSF criteria, there was an obvious selection bias for some important clinical characteristics in the LDLT group. Patients having LDLT for HCC had fewer incidental tumors, a lower rate of preoperative transarterial chemoembolization (TACE), a lower rate of salvage transplantation (with pretransplant resection or ablation), shorter waiting time on list, and a lower graft-weight-to-standard-liver-weight ratio. The inferior oncological outcomes in the LDLT group were possibly caused by more aggressive tumor behavior and small-for-size graft injury and regeneration[24]. Although the overall survival rates were comparable between the LDLT and DDLT groups, the cumulative 5-year HCC recurrence rate was significantly higher in LDLT group (29% vs 0%). Thus, selection of patients with early HCC based on standard tumor size and number for LDLT and DDLT may eventually result in different clinical outcomes. When considering a patient for an LDLT, besides a certain set of patient selection criteria, there are more factors to be taken into account, which include the unique nature of a living-donor graft as a dedicated gift to the recipient and potential donor risks, and additional clinical characteristics should also be considered and good preoperative counseling should be given to the donor and patient. In Hong Kong, the policy of “6-mo-wait” before salvage transplantation does not apply to LDLT, since both donors and recipients willingly accept the relatively higher recurrence rate with the realization that LDLT is their only option.

TREATMENTS FOR HCC RECURRENCE

Theoretically, all modalities for treating HCC can be used to treat its recurrence. Aggressive treatments can usually be given to patients who have satisfactory liver function and no widespread tumor cell dissemination. However, HCC recurrence after LT is considered a “systemic disease”, and the efficacy of locoregional treatment for a systemic disease is doubtful. For LT recipients, the use of immunosuppressants may hinder wound healing and thus lead to a higher chance of infective complications. Variable vascular anatomy in a graft liver or dense adhesion at the hilum may cause damage to important structures during dissection. Difficulties may be encountered in interventional radiological procedures like TACE when the catheter is negotiating through the arterial anastomosis. The use of targeted agents for post-LT HCC recurrence has not been validated by any large randomized trials and it may have adverse effects on immunocompromised patients. A multidisciplinary approach with the involvement of hepatologists, surgeons, radiologists, oncologists and radiation oncologists is definitely for the best interest of this group of patients.

Liver resection and local ablative therapy for intrahepatic recurrence

Catalano et al[25] reported the initial results of graft liver resection for graft ischemic damage in 12 patients. The perioperative mortality rate was high at 66.6%, manifesting the difficulty of graft liver resection in the presence of sepsis. On the other hand, Sommacale et al[26] reported that graft liver resection for intrahepatic recurrence achieved a low mortality rate and satisfactory long-term survival with a median follow-up of 92 mo. Nonetheless, there were only 3 patients in the series. According to unpublished data from the only LT center in Hong Kong, in 252 patients who underwent LT for HCC, 35 had disease recurrence. Three patients had only intrahepatic recurrence and underwent aggressive resection. This very small series had a 66.7% 3-year survival and 0% mortality. Actually, all reported series were small and the studies had a retrospective nature with significant selection biases. Hence, more evidence is needed to support graft liver resection as a good treatment for HCC recurrence.

Radiofrequency ablation (RFA), a local ablative treatment, is the established treatment option for resectable and unresectable HCCs. Its efficacy has been shown to be comparable to that of partial liver resection in treating small HCCs[27]. It would be reasonable to extrapolate that RFA can be an option for treating post-LT intrahepatic recurrence of HCC too. A case report showed that percutaneous RFA achieved 2-year disease-free survival in a 65-year-old patient who had a solitary recurrent HCC inside the graft liver[28].

Stereotactic body radiation therapy and intra-arterial infusion of yttrium-90 microspheres for intrahepatic recurrence

Numerous advances in external-beam radiation therapy have allowed more accurate targeting and made aggressive dose-fractionation strategies possible with techniques such as stereotactic body radiation therapy (SBRT). As a kind of radiosurgery, SBRT was originally developed to treat intracranial malignancies. It has since been adopted to treat extracranial diseases. The use of SBRT as treatment of HCC has yet to be established, but it is tested by a number of clinical trials for its efficacy in treating unresectable and unablatable HCCs. Initial results showed that it achieved a local control rate of 87%-100%[17,29-31].

Intra-arterial infusion of yttrium-90 microspheres (Y-90 SIR) is an established treatment for unresectable HCCs[32] and has gained popularity in recent years. It is often used to treat advanced HCC, especially in patients with a large tumor burden, suboptimal performance status, or lobar portal vein thrombosis[33]. Chan et al[34] reported that in the treatment of primary HCC, it achieved a 38%-65% partial response rate and a median survival duration of 23 mo, which is 2.6-4.7 times the duration seen in historic controls. In a recent study of 20 patients with unresectable HCCs, it achieved an overall survival rate of 90% at a median follow-up period of 275 d (range: 32-677 d)[33]. However, the data on the use of SBRT and intra-arterial infusion of Y-90 SIR for recurrent HCC after LT are extremely scarce. In the only two case reports, complete tumor necrosis was observed in a 52-year-old and a 42-year-old patient with solitary intrahepatic recurrence of HCC after a course of SBRT and intra-arterial infusion of Y-90 SIR respectively[35,36].

TACE for intrahepatic recurrence

TACE is often used as a bridging therapy for waitlisted patients and its results are satisfactory. Lo et al[37] reported that it resulted in marked tumor response, and the actuarial survival was significantly better in the TACE group (1 year: 57%, 2 years: 31%, 3 years: 26%) compared with the control group (1 year: 32%, 2 years: 11%, 3 years: 3%, P = 0.002). When adjustments for baseline variables that were prognostic on univariate analysis were made with a multivariate Cox model, the survival benefit of TACE remained significant (relative risk of death: 0.49; 95%CI: 0.29-0.81; P = 0.006).

Chok et al[38] compared TACE and RFA for unresectable HCCs and found that they were comparable in terms of time to disease progression (P = 0.95) and overall survival (P = 0.02).

Successful outcomes of TACE therapy (with and without the use of iodized oil) for the treatment of recurrent intrahepatic HCC after LT have been reported[39,40] although the studies were small and retrospective in nature. As said before, the transcatheter procedure can be technically demanding in the presence of distorted vasculature in a post-LT setting.

New therapy for intrahepatic recurrence

High-intensity focused ultrasound (HIFU) ablation is a relatively new totally extracorporeal treatment for unresectable HCCs. Ng et al[41] in their initial research reported that it achieved a primary effective treatment rate of 79.5% and 1-year and 3-year overall survival rates of 87.7% and 62.4% respectively.

Cheung et al[42] compared HIFU ablation with TACE and reported that HIFU ablation achieved rates of complete tumor response, partial tumor response, stable disease and progressive disease (in accordance with the modified Response Evaluation Criteria in Solid Tumors) of 50%, 7.7%, 25.6% and 7.7% respectively. As with TACE, the corresponding rates were 0%, 21.2%, 63.5% and 15.4% respectively (P < 0.0001). The 1-year, 3-year and 5-year survival rates achieved by HIFU ablation were 84.6%, 49.2% and 32.3% respectively, and those by TACE were 69.2%, 29.8% and 2.3% respectively (P = 0.001).

Chan et al[43] compared HIFU ablation with RFA in terms of survival. The two kinds of ablative treatment produced similar results. The 1-year, 2-year, and 3-year disease-free survival rates were 37.0%, 25.9% and 18.5% respectively in the HIFU group, and 48.6%, 32.1% and 26.5% respectively in the RFA group (P = 0.61). The 1-year, 2-year, and 3-year overall survival rates were 96.3%, 81.5% and 69.8% respectively in the former, and 92.1%, 76.1% and 64.2% respectively in the latter (P = 0.19).

In the pilot study on HIFU ablation as a bridging therapy for HCC patients waitlisted for LT conducted at the only LT center in Hong Kong, it was found that with the availability of HIFU ablation, the rate of receiving bridging therapy increased dramatically from 39.2% to 80.4%. HIFU ablation and TACE achieved similar percentages of tumor necrosis as seen in excised livers (P = 0.353), and both treatments resulted in significantly higher necrosis rates than that in the best medical treatment group (P = 0.010 and 0.020)[44]. As HIFU ablation has been shown to be a useful bridging therapy, it should have great potential in the management of recurrent HCC after LT.

Treatment for multiple recurrence

Mammalian target of rapamycin (mTOR) inhibitors have been shown to have a direct antitumorigenic effect and to be able to inhibit cell growth[45-47]. In experimental models of HCC, the mTOR pathway was aberrantly activated in up to half of the cases. Although the currently available data came from retrospective studies and are premature, there is the hope that mTOR-based immunosuppressive therapy after LT will one day come into use[48]. The use of sorafenib, an inhibitor of multiple tyrosine kinases (including c-Raf and b-Raf), has been approved as a first-line treatment for advanced HCC[49]. Activation of the Ras/mitogen-activated protein kinase pathway is a common finding in neoplastic processes (including in HCC) and is a determinant for promoting cell proliferation and the survival of tumor cells. This makes sorafenib an interesting drug; its use as a treatment for unresectable HCCs and as an adjuvant treatment before and after HCC recurrence is being investigated[50]. A study from Spain demonstrated that combination therapy resulted in an overall response (in accordance with the Response Evaluation Criteria in Solid Tumors) rate of 3.8% (1/26), and there was sustained stabilization of disease in 13 additional cases (50.0%)[42]. The median overall survival was 19.3 mo (95%CI: 13.4-25.1 mo), and the median time to progression was 6.77 mo (95%CI: 2.3-11.1 mo). Although a few studies have shown that there is some evidence of synergistic anticancer activity, early-phase clinical studies of mTOR inhibitors plus sorafenib for advanced HCC reported ambivalent findings, which were the results of increased toxicity (e.g., hand-foot syndrome) in combination therapy[51,52]. In a recent study from Italy, the outcomes of sorafenib treatment for post-LT HCC recurrence were significantly better than those of best medical care [median patient survival from recurrence: 21.3 mo vs 11.8 mo, hazard ratio (HR) = 5.2, P = 0.0009; median patient survival from untreatable presentation or progression: 10.6 mo vs 2.2 mo, HR = 21.1, P < 0.0001]. The only factor associated with survival found by multivariate analysis was treatment with sorafenib (HR = 4.0, P = 0.0325). No severe adverse event was registered[53]. Individualized treatment should be tailor-made for individual recipients, and input from oncologists would be of great value. However, drug toxicity is a major concern as shown in many studies, and their recommendations should not be overlooked.

Use of different immunosuppressants

It has been suggested that immunosuppressive therapy should switch from using non-mTOR inhibitors to using mTOR inhibitors. Another suggestion is that mTOR inhibitors can be used as an add-on. Monaco[54] found that the use of mTOR inhibitors might decrease the incidence of new malignancy after transplantation, mainly skin cancer.

A clinical trial by Alamo et al[55] comparing calcineurin inhibitors with everolimus and sirolimus for patients who received LT for oncological disease reported that the HCC recurrence rate was significantly lower and survival significantly prolonged in patients receiving either everolimus or sirolimus. A meta-analysis by Liang et al[56] endorsed the safety and efficacy of sirolimus-based immunosuppression for patients who received LT for HCC. Pooled results of the five studies eligible for evaluation showed that sirolimus-based regimens prolonged overall survival (OR = 2.47; 95%CI: 1.72-3.55) and decreased tumor recurrence (OR = 0.42; 95%CI: 0.21-0.83), with no significant differences in acute rejection and hepatic artery thrombosis.

A United States study compared sirolimus-based maintenance therapy with calcineurin inhibitor treatment for recipients of LT for HCC and found that overall survival was better in the sirolimus arm[57]. Clinical trials examining the anticancer effects of mTOR inhibitors in recipients of LT for HCC have shown encouraging results[58]. On multivariate analysis in a large Canadian trial, sirolimus-based maintenance therapy was one of the factors associated with improved survival after LT for HCC (HR = 0.53, 95%CI: 0.31-0.92, P≤ 0.05)[59].

The reported results of using these relatively new agents has suggested that they may prevent or reduce the incidence of HCC recurrence after LT, but a definite answer from large randomized controlled trials is still lacking.

CONCLUSION

Recurrence of HCC after LT is a deadly disease. Although there are a variety of treatment approaches, long-term cure is rarely seen. One of the reasons is that the disease is “systemic” in most of the cases, even if the recurrence is intrahepatic only. Effective adjuvant or systemic therapy has yet to be identified. A multidisciplinary approach with fine-tuning of treatment goals and objectives will definitely be beneficial, and development of new drugs or modification of current systemic agents is urgently needed.

Footnotes

P- Reviewer: Dehghani SM, Frenette C, Kubota K, Silva G

S- Editor: Ji FF L- Editor: A E- Editor: Liu SQ

References
1.  Bruix J, Sherman M, Llovet JM, Beaugrand M, Lencioni R, Burroughs AK, Christensen E, Pagliaro L, Colombo M, Rodés J. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. European Association for the Study of the Liver. J Hepatol. 2001;35:421-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3252]  [Cited by in F6Publishing: 3169]  [Article Influence: 137.8]  [Reference Citation Analysis (0)]
2.  El-Serag HB, Davila JA, Petersen NJ, McGlynn KA. The continuing increase in the incidence of hepatocellular carcinoma in the United States: an update. Ann Intern Med. 2003;139:817-823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 714]  [Cited by in F6Publishing: 758]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
3.  Welker MW, Bechstein WO, Zeuzem S, Trojan J. Recurrent hepatocellular carcinoma after liver transplantation - an emerging clinical challenge. Transpl Int. 2013;26:109-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
4.  Luan FL, Hojo M, Maluccio M, Yamaji K, Suthanthiran M. Rapamycin blocks tumor progression: unlinking immunosuppression from antitumor efficacy. Transplantation. 2002;73:1565-1572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 193]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
5.  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)]
6.  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)]
7.  Ng KK, Lo CM, Chan SC, Chok KS, Cheung TT, Fan ST. Liver transplantation for hepatocellular carcinoma: the Hong Kong experience. J Hepatobiliary Pancreat Sci. 2010;17:548-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
8.  El-Serag HB, Mason AC. Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med. 1999;340:745-750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2221]  [Cited by in F6Publishing: 2124]  [Article Influence: 85.0]  [Reference Citation Analysis (0)]
9.  Taura N, Yatsuhashi H, Hamasaki K, Nakao K, Daikoku M, Ueki T, Yano K, Matsumoto T, Ishibashi H, Eguchi K. Increasing hepatitis C virus-associated hepatocellular carcinoma mortality and aging: Long term trends in Japan. Hepatol Res. 2006;34:130-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
10.  Chok KS, Chan SC, Cheung TT, Chan AC, Fan ST, Lo CM. Late recurrence of hepatocellular carcinoma after liver transplantation. World J Surg. 2011;35:2058-2062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
11.  Chan SC, Sharr WW, Chok KS, Chan AC, Lo CM. Wait and transplant for stage 2 hepatocellular carcinoma with deceased-donor liver grafts. Transplantation. 2013;96:995-999.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
12.  Kornberg A, Küpper B, Tannapfel A, Katenkamp K, Thrum K, Habrecht O, Wilberg J. Long-term survival after recurrent hepatocellular carcinoma in liver transplant patients: clinical patterns and outcome variables. Eur J Surg Oncol. 2010;36:275-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 98]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
13.  Sotiropoulos GC, Molmenti EP, Lösch C, Beckebaum S, Broelsch CE, Lang H. Meta-analysis of tumor recurrence after liver transplantation for hepatocellular carcinoma based on 1,198 cases. Eur J Med Res. 2007;12:527-534.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  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)]
15.  Chinnakotla S, Davis GL, Vasani S, Kim P, Tomiyama K, Sanchez E, Onaca N, Goldstein R, Levy M, Klintmalm GB. Impact of sirolimus on the recurrence of hepatocellular carcinoma after liver transplantation. Liver Transpl. 2009;15:1834-1842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 124]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
16.  Saborido BP, Díaz JC, de Los Galanes SJ, Segurola CL, de Usera MA, Garrido MD, Elola-Olaso AM, Sánz RG, Romero CJ, Garcia García I. Does preoperative fine needle aspiration-biopsy produce tumor recurrence in patients following liver transplantation for hepatocellular carcinoma? Transplant Proc. 2005;37:3874-3877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 49]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
17.  Andolino DL, Johnson CS, Maluccio M, Kwo P, Tector AJ, Zook J, Johnstone PA, Cardenes HR. Stereotactic body radiotherapy for primary hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2011;81:e447-e453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 257]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
18.  Cheung TT, Ho CL, Lo CM, Chen S, Chan SC, Chok KS, Fung JY, Yan Chan AC, Sharr W, Yau T. 11C-acetate and 18F-FDG PET/CT for clinical staging and selection of patients with hepatocellular carcinoma for liver transplantation on the basis of Milan criteria: surgeon’s perspective. J Nucl Med. 2013;54:192-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (1)]
19.  Cheung TT, Chan SC, Ho CL, Chok KS, Chan AC, Sharr WW, Ng KK, Poon RT, Lo CM, Fan ST. Can positron emission tomography with the dual tracers [11 C]acetate and [18 F]fludeoxyglucose predict microvascular invasion in hepatocellular carcinoma? Liver Transpl. 2011;17:1218-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 66]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
20.  de Villa V, Lo CM. Liver transplantation for hepatocellular carcinoma in Asia. Oncologist. 2007;12:1321-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
21.  Roayaie S, Schwartz JD, Sung MW, Emre SH, Miller CM, Gondolesi GE, Krieger NR, Schwartz ME. Recurrence of hepatocellular carcinoma after liver transplant: patterns and prognosis. Liver Transpl. 2004;10:534-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 344]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
22.  Fisher RA, Kulik LM, Freise CE, Lok AS, Shearon TH, Brown RS, Ghobrial RM, Fair JH, Olthoff KM, Kam I. Hepatocellular carcinoma recurrence and death following living and deceased donor liver transplantation. Am J Transplant. 2007;7:1601-1608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 190]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
23.  Lo CM, Fan ST, Liu CL, Chan SC, Ng IO, Wong J. Living donor versus deceased donor liver transplantation for early irresectable hepatocellular carcinoma. Br J Surg. 2007;94:78-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 185]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
24.  Man K, Fan ST, Lo CM, Liu CL, Fung PC, Liang TB, Lee TK, Tsui SH, Ng IO, Zhang ZW. Graft injury in relation to graft size in right lobe live donor liver transplantation: a study of hepatic sinusoidal injury in correlation with portal hemodynamics and intragraft gene expression. Ann Surg. 2003;237:256-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 144]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
25.  Catalano G, Urbani L, Biancofiore G, Bindi L, Boldrini A, Consani G, Bisà M, Campatelli A, Petruzzi P, Cioni R. Hepatic resection after liver transplantation as a graft-saving procedure: indication criteria, timing and outcome. Transplant Proc. 2004;36:545-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
26.  Sommacale D, Dondero F, Sauvanet A, Francoz C, Durand F, Farges O, Kianmanesh R, Belghiti J. Liver resection in transplanted patients: a single-center Western experience. Transplant Proc. 2013;45:2726-2728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Chen MS, Li JQ, Zheng Y, Guo RP, Liang HH, Zhang YQ, Lin XJ, Lau WY. A prospective randomized trial comparing percutaneous local ablative therapy and partial hepatectomy for small hepatocellular carcinoma. Ann Surg. 2006;243:321-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1100]  [Cited by in F6Publishing: 1039]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
28.  Ho CK, Chapman WC, Brown DB. Radiofrequency ablation of recurrent hepatocellular carcinoma in a patient after liver transplantation: two-year follow-up. J Vasc Interv Radiol. 2007;18:1451-1453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
29.  Cárdenes HR, Price TR, Perkins SM, Maluccio M, Kwo P, Breen TE, Henderson MA, Schefter TE, Tudor K, Deluca J. Phase I feasibility trial of stereotactic body radiation therapy for primary hepatocellular carcinoma. Clin Transl Oncol. 2010;12:218-225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 184]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
30.  Bujold A, Massey CA, Kim JJ, Brierley J, Cho C, Wong RK, Dinniwell RE, Kassam Z, Ringash J, Cummings B. Sequential phase I and II trials of stereotactic body radiotherapy for locally advanced hepatocellular carcinoma. J Clin Oncol. 2013;31:1631-1639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 508]  [Cited by in F6Publishing: 552]  [Article Influence: 50.2]  [Reference Citation Analysis (0)]
31.  Kang JK, Kim MS, Cho CK, Yang KM, Yoo HJ, Kim JH, Bae SH, Jung da H, Kim KB, Lee DH. Stereotactic body radiation therapy for inoperable hepatocellular carcinoma as a local salvage treatment after incomplete transarterial chemoembolization. Cancer. 2012;118:5424-5431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 244]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
32.  Benson AB, Abrams TA, Ben-Josef E, Bloomston PM, Botha JF, Clary BM, Covey A, Curley SA, D’Angelica MI, Davila R. NCCN clinical practice guidelines in oncology: hepatobiliary cancers. J Natl Compr Canc Netw. 2009;7:350-391.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Padia SA, Kwan SW, Roudsari B, Monsky WL, Coveler A, Harris WP. Superselective yttrium-90 radioembolization for hepatocellular carcinoma yields high response rates with minimal toxicity. J Vasc Interv Radiol. 2014;25:1067-1073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
34.  Chan AO, Yuen MF, Hui CK, Tso WK, Lai CL. A prospective study regarding the complications of transcatheter intraarterial lipiodol chemoembolization in patients with hepatocellular carcinoma. Cancer. 2002;94:1747-1752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 189]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
35.  Rivera L, Giap H, Miller W, Fisher J, Hillebrand DJ, Marsh C, Schaffer RL. Hepatic intra-arterial infusion of yttrium-90 microspheres in the treatment of recurrent hepatocellular carcinoma after liver transplantation: a case report. World J Gastroenterol. 2006;12:5729-5732.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Mazloom A, Hezel AF, Katz AW. Stereotactic body radiation therapy as a bridge to transplantation and for recurrent disease in the transplanted liver of a patient with hepatocellular carcinoma. Case Rep Oncol. 2014;7:18-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
37.  Lo CM, Ngan H, Tso WK, Liu CL, Lam CM, Poon RT, Fan ST, Wong J. Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology. 2002;35:1164-1171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1904]  [Cited by in F6Publishing: 1903]  [Article Influence: 86.5]  [Reference Citation Analysis (0)]
38.  Chok KS, Ng KK, Poon RT, Lam CM, Yuen J, Tso WK, Fan ST. Comparable survival in patients with unresectable hepatocellular carcinoma treated by radiofrequency ablation or transarterial chemoembolization. Arch Surg. 2006;141:1231-1236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
39.  Ko HK, Ko GY, Yoon HK, Sung KB. Tumor response to transcatheter arterial chemoembolization in recurrent hepatocellular carcinoma after living donor liver transplantation. Korean J Radiol. 2009;8:320-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
40.  Zhou B, Shan H, Zhu KS, Jiang ZB, Guan SH, Meng XC, Zeng XC. Chemoembolization with lobaplatin mixed with iodized oil for unresectable recurrent hepatocellular carcinoma after orthotopic liver transplantation. J Vasc Interv Radiol. 2010;21:333-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Ng KK, Poon RT, Chan SC, Chok KS, Cheung TT, Tung H, Chu F, Tso WK, Yu WC, Lo CM. High-intensity focused ultrasound for hepatocellular carcinoma: a single-center experience. Ann Surg. 2011;253:981-987.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 94]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
42.  Cheung TT, Poon RT, Jenkins CR, Chu FS, Chok KS, Chan AC, Tsang SH, Dai WC, Yau TC, Chan SC. Survival analysis of high-intensity focused ultrasound therapy vs. transarterial chemoembolization for unresectable hepatocellular carcinomas. Liver Int. 2014;34:e136-e143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
43.  Chan AC, Cheung TT, Fan ST, Chok KS, Chan SC, Poon RT, Lo CM. Survival analysis of high-intensity focused ultrasound therapy versus radiofrequency ablation in the treatment of recurrent hepatocellular carcinoma. Ann Surg. 2013;257:686-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
44.  Chok KS, Cheung TT, Lo RC, Chu FS, Tsang SH, Chan AC, Sharr WW, Fung JY, Dai WC, Chan SC. Pilot study of high-intensity focused ultrasound ablation as a bridging therapy for hepatocellular carcinoma patients wait-listed for liver transplantation. Liver Transpl. 2014;20:912-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
45.  Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy. Nat Rev Cancer. 2004;4:335-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1042]  [Cited by in F6Publishing: 1022]  [Article Influence: 51.1]  [Reference Citation Analysis (0)]
46.  Koehl GE, Andrassy J, Guba M, Richter S, Kroemer A, Scherer MN, Steinbauer M, Graeb C, Schlitt HJ, Jauch KW. Rapamycin protects allografts from rejection while simultaneously attacking tumors in immunosuppressed mice. Transplantation. 2004;77:1319-1326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 182]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
47.  Guba M, von Breitenbuch P, Steinbauer M, Koehl G, Flegel S, Hornung M, Bruns CJ, Zuelke C, Farkas S, Anthuber M. Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med. 2002;8:128-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1347]  [Cited by in F6Publishing: 1270]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
48.  Gomez-Martin C, Bustamante J, Castroagudin JF, Salcedo M, Garralda E, Testillano M, Herrero I, Matilla A, Sangro B. Efficacy and safety of sorafenib in combination with mammalian target of rapamycin inhibitors for recurrent hepatocellular carcinoma after liver transplantation. Liver Transpl. 2012;18:45-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 119]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
49.  Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9016]  [Cited by in F6Publishing: 9500]  [Article Influence: 593.8]  [Reference Citation Analysis (1)]
50.  Kudo M. Adjuvant therapy after curative treatment for hepatocellular carcinoma. Oncology. 2011;81 Suppl 1:50-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
51.  Finn RS, Poon RT, Yau T, Klümpen HJ, Chen LT, Kang YK, Kim TY, Gomez-Martin C, Rodriguez-Lope C, Kunz T. Phase I study investigating everolimus combined with sorafenib in patients with advanced hepatocellular carcinoma. J Hepatol. 2013;59:1271-1277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 61]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
52.  Kelley RK, Nimeiri HS, Munster PN, Vergo MT, Huang Y, Li CM, Hwang J, Mulcahy MF, Yeh BM, Kuhn P. Temsirolimus combined with sorafenib in hepatocellular carcinoma: a phase I dose-finding trial with pharmacokinetic and biomarker correlates. Ann Oncol. 2013;24:1900-1907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
53.  Sposito C, Mariani L, Germini A, Flores Reyes M, Bongini M, Grossi G, Bhoori S, Mazzaferro V. Comparative efficacy of sorafenib versus best supportive care in recurrent hepatocellular carcinoma after liver transplantation: a case-control study. J Hepatol. 2013;59:59-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 109]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
54.  Monaco AP. The role of mTOR inhibitors in the management of posttransplant malignancy. Transplantation. 2009;87:157-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 95]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
55.  Alamo JM, Bernal C, Marín LM, Suárez G, Serrano J, Barrera L, Sousa JM, Padillo FJ, Gómez-Bravo MA. Antitumor efficacy of mammalian target of rapamycin inhibitor therapy in liver transplant recipients with oncological disease: a case-control study. Transplant Proc. 2012;44:2089-2092.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
56.  Liang W, Wang D, Ling X, Kao AA, Kong Y, Shang Y, Guo Z, He X. Sirolimus-based immunosuppression in liver transplantation for hepatocellular carcinoma: a meta-analysis. Liver Transpl. 2012;18:62-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 129]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
57.  Zimmerman MA, Trotter JF, Wachs M, Bak T, Campsen J, Skibba A, Kam I. Sirolimus-based immunosuppression following liver transplantation for hepatocellular carcinoma. Liver Transpl. 2008;14:633-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 174]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
58.  Toso C, Meeberg GA, Bigam DL, Oberholzer J, Shapiro AM, Gutfreund K, Ma MM, Mason AL, Wong WW, Bain VG. De novo sirolimus-based immunosuppression after liver transplantation for hepatocellular carcinoma: long-term outcomes and side effects. Transplantation. 2007;83:1162-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 153]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
59.  Toso C, Merani S, Bigam DL, Shapiro AM, Kneteman NM. Sirolimus-based immunosuppression is associated with increased survival after liver transplantation for hepatocellular carcinoma. Hepatology. 2010;51:1237-1243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 237]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]