Predisposing factors for hepatocellular carcinoma recurrence following initial remission after transcatheter arterial chemoembolization

  • Authors:
    • Akitoshi Douhara
    • Tadashi Namisaki
    • Kei Moriya
    • Mitsuteru Kitade
    • Kosuke Kaji
    • Hideto Kawaratani
    • Kosuke Takeda
    • Yasushi Okura
    • Hiroaki Takaya
    • Ryuichi Noguchi
    • Norihisa Nishimura
    • Kenichiro Seki
    • Shinya Sato
    • Yasuhiko Sawada
    • Junichi Yamao
    • Akira Mitoro
    • Masakazu Uejima
    • Tsuyoshi Mashitani
    • Naotaka Shimozato
    • Soichiro Saikawa
    • Keisuke Nakanishi
    • Masanori Furukawa
    • Takuya Kubo
    • Hitoshi Yoshiji
  • View Affiliations

  • Published online on: June 28, 2017     https://doi.org/10.3892/ol.2017.6489
  • Pages: 3028-3034
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is prone to recurrence following curative treatment. The purpose of the present study was to identify the predisposing factors of HCC recurrence following complete remission achieved by transarterial chemoembolization (TACE). A retrospective cohort study of 70 consecutive patients with HCC who underwent TACE as the initial treatment was conducted. The patients were divided into two groups according to their 1‑year disease‑free survival (DFS) status; the early recurrence group (ER group; n=32), with HCC recurring within 1 year of initial TACE; and the non‑early recurrence group (NER group; n=38), who did not experience recurrence within 1 year. The parameters identified as significantly associated with DFS time on univariate analysis were aspartate aminotransferase (AST), alanine aminotransferase and α‑fetoprotein levels, as well as the tumor number (P=0.003, P=0.027, P=0.002 and P=0.005, respectively). Multivariate analysis revealed that AST levels and tumor number were significantly associated with a shorter DFS period (P=0.009 and P=0.038, respectively). The Mantel‑Haenszel test revealed a significant trend of decreasing DFS with increasing tumor number. Among the patients with HCC in the ER group, locoregional recurrence occurred more frequently in those who received TACE alone compared with those treated with TACE combined with radiofrequency ablation treatment. In summary, multinodularity of HCC is the most potent predictive factor for the recurrence of HCC within 1 year of initial TACE.

Introduction

Hepatocellular carcinoma (HCC) is the sixth most common type of cancer worldwide (1), and develops in patients with chronic liver disease and cirrhosis (2). The treatment of HCC depends on its stage of progression (3). Surgical resection remains the mainstay of potentially curative therapy, but the majority of patients with HCC have passed the opportunity for treatment with surgery at the time of diagnosis (4,5).

Transcatheter arterial chemoembolization (TACE) is an essential treatment option for patients with HCC who cannot be treated with other potentially more effective therapies, including surgical resection or local ablative therapies, and is the standard of care for patients with noninvasive multinodular tumors at an intermediate stage (6,7). However, the long-term outcomes of patients treated with TACE are not entirely satisfactory, mainly due to the low tumor necrosis rates (8) and high recurrence rates (6).

Radiofrequency (RF) ablation (RFA) represents a safe and effective first-line locoregional treatment for HCCs with three tumors or fewer, of ≤3 cm in size (3,9). Despite the high complete necrosis rate of RFA, early local or distant tumor recurrence within 1 year may still occur (10). TACE combined with RFA can enhance the advantages of each individual treatment (8) and increase their cooperative effect, demonstrating the potential benefits of a multidisciplinary approach for advanced HCC (11,12). Furthermore, the combination therapy of RFA and TACE has been shown to be superior to TACE or RFA alone in increasing locoregional control and improving the curative effect and survival time in patients with advanced HCC (13,14). However, the risk factors for tumor recurrence following treatment of HCC have not yet been clarified in detail.

The aims of the present study were to identify the characteristics of HCC associated with recurrence following successful initial treatment with TACE, and to compare the recurrence patterns between patients with HCC who received TACE alone and those treated with TACE combined with RFA treatment.

Methods and materials

Study design

The present study was a retrospective cohort study performed at a single center. The medical records of 357 patients treated with TACE between June 2009 and June 2013 at Nara Medical University (Kashihara, Japan) were reviewed (Fig. 1). Of these, 70 patients were initially treated with TACE and subsequently observed over a 1-year period. These patients were divided into two groups according to DFS status at 1 year: The early recurrence (ER) group (recurrence within 1 year after initial TACE; n=32) and the non-early recurrence (NER) group (no recurrence within 1 year after initial TACE; n=38). Of the 32 patients in the ER group, 5 did not achieve a complete remission (CR), with 2 succumbing to HCC. Of the 38 patients in the NER group, 1 succumbed to HCC, while 15 did not experience a recurrence of HCC for >1 year after the initial TACE. The degree of lipiodol retention in the tumor within 1 week of TACE was routinely evaluated by multi-detector row computed tomography (MDCT). RFA was administered if MDCT and contract-enhanced ultrasonography (CE-US) detected a residual viable tumor. The study was approved by the local ethics committee of Nara Medical University (Nara, Japan) and written informed consent was obtained from all patients prior to treatment.

Diagnosis of HCC

The diagnosis of HCC was confirmed without biopsy in patients with chronic liver disease and cirrhosis who had a tumor that exhibited a typical vascular pattern on dynamic imaging modalities [such as contrast-enhanced MDCT (CE-MDCT) and gadolinium ethoxybenzyl diethylenetriamine pentaacetic acid (Gd-EOB-DTPA)-enhanced magnetic resonance imaging (MRI)], in accordance with the practice guidelines of the Japan Society of Hepatology (15). However, if the vascular profile on imaging was not characteristic, or if a nodule was detected in a healthy normal liver, supplementary tests, including Gd-EOB-DTPA-enhanced MRI, CE-US, computed tomography (CT) angiography and liver tumor biopsy, were considered. All patients were examined by CE-MDCT or Gd-EOB-DTPA-enhanced MRI every 2 or 3 months after initial TACE.

TACE

The procedure for TACE has previously been described in detail (16). Briefly, a single femoral approach was used, following Seldinger's technique (17), by inserting a 4-Fr catheter (RH-6SP0061i; Terumo Medical Corporation, Tokyo, Japan) over a 5-Fr introducer sheath into the celiac artery, using superior mesenteric artery angiography as well as selective hepatic arteriography to identify tumor feeders. The artery was selectively catheterized with a microcatheter/microguidewire system and embolized with a mixture of epirubicin with iodized oil (lipiodol; Laboratoire Andre Guerbet, Aulnay-sous-Bois, France). The feeders were then embolized with gelatin sponge pledgets (Cutanplast; Mascia Brunelli S.p.A, Milan, Italy) until complete stasis of the blood flow was detected by angiography. Collateral artery embolization was performed if branches such as the phrenic artery and internal thoracic artery were engaged in the tumor blood supply. Post-TACE cone-beam CT was performed to assess the extent of lipiodol uptake in the tumor at the end of TACE.

Percutaneous RFA

RFA of HCC was performed using the Cool-tip™ RF system (Integra Burlington MA, Inc., MA, USA). RFA with ultrasound guidance was conducted under general and local anesthesia, using a 3.5-MHz probe with an incorporated guide and a 17-gauge cooled-tip electrode (Cool-tip; Valleylab, Burlington, MA, USA) with a 2- or 3-cm exposed portion. This system consisted of an RF generator to produce a current of 480 kHz at a maximal power of 200 W, a single-electrode RFA, a water-pumping machine and return grounding pads. The RFA started at a low power (40 or 60 W) and increased by 10 W/min, and the delivery of RF energy was automatically modulated according to the tissue impedance around the electrode. Tumor ablation continued at maximum power until the tissue impedance increased to the point at which the power output fell rapidly (the ‘break’). The treatment response was assessed based on CE-MDCT or CE-US performed within 1 week of RFA treatment.

Assessment and follow-up

The Response Evaluation Criteria in Solid Tumors (RECIST) criteria (18) are recommended for evaluating treatment efficacy in clinical trials and practice. The response to treatment was assessed by CE-MDCT according to the RECIST criteria at 4 weeks post-TACE or within 1 week of RFA, and additional RFA was performed until no residual viable tumor was detectable. Following the final treatment session, patients were evaluated every 3 months for 2 years and followed up every 6 months thereafter by CE-MDCT or CE-MRI. The primary end-point was the HCC DFS period following initial TACE.

Statistical analysis

Categorical variables were analyzed using the Mantel-Haenszel test. Bivariate analyses of nominal parameters were performed using the χ2 test. Univariate and multivariate logistic regression analyses were conducted to assess the effect of different parameters on HCC recurrence following initial TACE. Data are presented as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference. All statistical analyses were performed using IBM SPSS statistics version 22 (IBM Corp., Armonk, NY, USA).

Results

Clinical characteristics of patients

Baseline characteristics of the enrolled 70 patients are summarized in Table I. The patient group included 51 males (73%) and 19 females (27%), and the mean age was 70.5±8.9 years. A total of 54 patients (77%) were classified as having Child-Pugh class (19) A cirrhosis, and 16 (23%) patients were classified as having class B cirrhosis. At the initial diagnosis, the mean tumor number was 2.3±2.1, and 8 patients (11%) had multiple lesions. The mean serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were 53.3±43.4 and 40.3±36.4 IU/l, respectively. The mean α-fetoprotein (AFP) level was 1,450.0±7,430.0 ng/ml.

Table I.

Baseline characteristics of the patients (n=70).

Table I.

Baseline characteristics of the patients (n=70).

VariableValue
Age, yearsa70.5±8.9 (49–83)
Sex, n
  Male51
  Female19
Etiology, n
  HBV12
  HCV37
  Others21
Child-Pugh classification, n
  A54
  B16
TNM stage, n
  I20
  II29
  III19
  IV2
Tumor size, cma3.4±3.1 (0.7–20.0)
Tumor numbera2.3±2.1 (1–9)
Aspartate transaminase, IU/la53.3±43.4 (12–285)
Alanine aminotransferase, IU/la40.3±36.4 (11–236)
α-fetoprotein, ng/mla 1,450.0±7,430.0
(1.7–56,831.6)
Protein induced by vitamin K 3,342.0±10,537.0
absence/antagonist-II, mAU/mla(7.0–66,753.0)

a Mean ± standard deviation (range). TNM, tumor-node-metastasis; HBV, hepatitis B virus; HCV, hepatitis C virus.

Risk factors associated with HCC recurrence

Univariate and multivariate analyses were performed to identify predictive risk factors for HCC recurrence through comparisons between the ER and NER groups. Univariate analysis revealed that the levels of AST, ALT and AFP, as well as tumor number, were associated with early HCC recurrence following TACE (Table II). The ER and NER groups did not differ significantly in terms of mean age, sex ratio, tumor etiology, Child-Pugh classification, tumor stage, mean tumor size or levels of protein induced by vitamin K absence/antagonist-II. Multivariate logistic regression analysis indicated that AST levels [odds ratio (OR), 1.069; P=0.009] and tumor number (OR, 1.661; P=0.038) were independent risk factors associated with HCC recurrence following initial TACE (Table III).

Table II.

Baseline characteristics of the patients.

Table II.

Baseline characteristics of the patients.

VariableEarly recurrence group (n=32)Non-early recurrence group (n=38) P-valuea
Age, yearsb69.5±9.0 (50–83)71.3±8.8 (49–82)0.354
Sex, n 0.865
  Male2328
  Female910
Etiology, n 0.871
  HBV57
  HCV1819
  Others912
Child-Pugh classification, n 0.695
  A2430
  B88
TNM stage, n 0.555
  I713
  II1316
  III118
  IV11
Tumor size, cmb4.4±4.2 (1.7–20.0)2.6±1.3 (0.7–6.6)0.153
Tumor numberb3.0±2.7 (1–9)1.6±0.9 (1–4)0.005
Aspartate transaminase, IU/lb69.8±57.4 (24–285)39.4±18.1 (12–96)0.003
Alanine aminotransferase, IU/lb50.2±46.2 (11–236)32.1±22.9 (13–126)0.027
α-fetoprotein, ng/mlb3144.6±10,836.0 (2.3–56,831.6)22.7±35.2 (1.7–144.2)0.002
Protein induced by vitamin K absence/antagonist-II, mAU/mlb6,396.5±14,871.0 (7.0–66,753.0)646.9±1,291.0 (8.0–5,845.0)0.053

a P-values represent comparisons between the early recurrence group and the non-early recurrence group.

b Mean ± standard deviation (range). TNM, tumor-node-metastasis; HBV, hepatitis B virus; HCV, hepatitis C virus.

Table III.

Multiple logistic regression analysis of risk factors for recurrence within 1 year of initial transcatheter arterial chemoembolization for hepatocellular carcinoma.

Table III.

Multiple logistic regression analysis of risk factors for recurrence within 1 year of initial transcatheter arterial chemoembolization for hepatocellular carcinoma.

VariableP-valueOdds ratio95% confidence interval
Aspartate transaminase0.0091.0691.017–1.125
Alanine aminotransferase0.2200.9160.270–1.001
α-fetoprotein0.1422.9610.740–6.372
Tumor number0.0381.6611.029–2.680
Association between tumor number and recurrence patterns with DFS period following initial TACE

Of the 70 patients, 32 had a single nodule, 20 had two nodules, 7 had three nodules and 11 had four or more nodules. The mean HCC DFS periods for patients with one, two, three and four or more nodules were 19.8, 13.4, 9.1 and 7.4 months, respectively. The χ2 test for trend demonstrated an inverse association between tumor number and DFS period in these patients (Fig. 2). Recurrence within 1 year of TACE was documented in 27 patients, including distant recurrence in 11 (41%) of these patients, and locoregional recurrence in 16 (59%) patients. Recurrence after 1 year was documented in 22 patients, including distant recurrence in 17 (77%) of these patients and locoregional recurrence in 5 (23%) patients. The association between recurrence patterns of HCC and DFS following initial TACE was also analyzed. A χ2 test revealed that, among all patients with recurrence, the proportions of locoregional and distant recurrence differed significantly between the ER group and the NER group (P<0.05), with a greater proportion of regional recurrence observed in the ER group (Fig. 3).

Comparison of HCC recurrence patterns between combined TACE and RFA treatment and TACE alone

In the ER group (n=32), 5 patients underwent TACE and RFA and the remaining 27 patients received TACE alone. In the NER group, all 38 patients received TACE alone. No locoregional recurrence was observed following treatment with a combination of TACE and RFA. Of the 11 patients who experienced distant recurrence in the ER group, 7 (64%) patients had undergone a combination of TACE and RFA and 4 (36%) patients received TACE alone. A χ2 test revealed that the combination treatment of TACE and RFA was associated with a significantly lower locoregional recurrence rate compared with TACE treatment alone in patients of the ER group (P<0.05; Fig. 4). Patients treated with TACE alone had a higher incidence of distant recurrence compared with those treated with combination therapy.

Discussion

TACE is a well-established procedure that offers a palliative survival benefit for patients with HCC that is unresectable or not suitable for local ablative treatment (20). Several case-control and retrospective studies have revealed a benefit of TACE for patient survival when comparing TACE-treated patients with untreated or historical controls (the patients with unresectable HCC received conservative treatment) (9,20). It has been a challenge for patients with advanced HCC to maintain a CR following TACE due to extracapsular invasion of HCC and residual viable cancer cells around the fibrous capsules subsequent to TACE (21). Complications associated with TACE, including an impaired hepatic functional reserve, support the use of RFA rather than repeated TACE treatments (22,23). Emerging evidence suggests that a combination of TACE and RFA exerts a synergistic anticancer activity against HCC, particularly for larger lesions that do not respond sufficiently to either TACE or RFA treatments alone (2426). An analysis of the factors that carry a high risk of early recurrence following TACE may improve the selection of patients suited to a combination of TACE with RFA.

In the present study, the clinical courses of 70 patients with HCC treated with TACE, and the different risk factors associated with HCC recurrence following initial remission after TACE, were examined. To the best of our knowledge, this is the first study to show that tumor number is the most important risk factor for recurrence within 1 year of initial TACE. The findings of the present study demonstrated an inverse association between tumor number and DFS time following initial TACE. Consistent with the present study, several previous studies have identified tumor number as a predictor of intrahepatic recurrence following initial TACE for HCC (2731). Recurrence following initial remission by TACE has been more often reported in patients with multinodular-type HCC and with portal vein thrombosis (32). By contrast, Matsuda et al (33) conjectured that tumor multiplicity was not associated with 1-year HCC recurrence status. It has also been reported that the risk factors for early HCC recurrence subsequent to achieving CR by TACE included large tumor size, non-compact lipiodol uptake and an AFP concentration >20 ng/ml, but not tumor number (34). This discrepancy may be explained in part by the different treatment modalities and the baseline characteristics of the patients with HCC between studies. HCC often consists of different cell types, including moderately differentiated and undifferentiated carcinoma (35,36), which is not eligible for TACE due to its hypovascularity. Early recurrence following a CR achieved by TACE may be mainly attributed to residual tumors that were undetectable on angiography. The distribution of lipiodol uptake determined by tumor differentiation and the blood vessel network have been shown to affect the local recurrence rate and long-term outcome in patients with HCC (37,38). Furthermore, consistent with the present study, a previous study showed that local recurrence developed more frequently in patients with early recurrence (≤1 year) compared with those with late recurrence (>1 year) (27). These results indicated that early recurrence is associated with local recurrence arising from limitations of the radiological evaluation of tumor response, and remnant tumors can develop and be recognizable by imaging modalities over time.

In combined treatment with TACE and RFA, the main roles of TACE are to counteract the heat-sink effect of hepatic blood flow by hepatic artery embolization (39) and to reduce the portal venous flow by filling the peripheral portal vein around the HCC (40). TACE can also lead to ischemic edema, which may enlarge the area of tumor necrosis induced by RFA (24). The combination of TACE and RFA generally results in complete tumor remission if the liver function reserve is sufficiently maintained post-TACE (12). No local HCC recurrence was observed in patients treated with a combination of TACE and RFA in the present study. The combination of TACE and RFA has significant advantages in terms of local tumor control and longer patient survival compared with TACE alone (41,42). These findings reinforce the notion that the combination of TACE and RFA resulted in a more efficient microscopic local tumor response compared with TACE alone.

The present study had a number of limitations. First, it was conducted in a single center and the sample size of patients with HCC recurrence was small. Second, there may have been unexpected factors that effected the probability of therapeutic response and recurrence. Third, patients who were lost to follow-up within 1 year and those who succumbed to the disease were excluded from the analysis, since the presence of early recurrence could not be confirmed; this may have led to selection bias.

In conclusion, multinodularity of HCC is an independent risk factor for 1-year recurrence of HCC in patients with initial remission following TACE. In particular, in patients who developed early recurrence, intrahepatic local recurrence more frequently occurred in HCC patients who received TACE alone compared with those treated with TACE combined with RFA treatment, which indicated that the combination treatment of TACE and RFA may be beneficial in preventing early recurrence of HCC.

References

1 

Forner A, Llovet JM and Bruix J: Hepatocellular carcinoma. Lancet. 379:1245–1255. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Simonetti RG, Cammà C, Fiorello F, Politi F, D'Amico G and Pagliaro L: Hepatocellular carcinoma. A worldwide problem and the major risk factors. Dig Dis Sci. 36:962–972. 1991. View Article : Google Scholar : PubMed/NCBI

3 

Bruix J and Sherman M: American Association for the Study of Liver Diseases: Management of hepatocellular carcinoma: An update. Hepatology. 53:1020–1022. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Hwang S, Lee SG, Ko GY, Kim BS, Sung KB, Kim MH, Lee SK and Hong HN: Sequential preoperative ipsilateral hepatic vein embolization after portal vein embolization to induce further liver regeneration in patients with hepatobiliary malignancy. Ann Surg. 249:608–616. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Ribero D, Abdalla EK, Madoff DC, Donadon M, Loyer EM and Vauthey JN: Portal vein embolization before major hepatectomy and its effects on regeneration, resectability and outcome. Br J Surg. 94:1386–1394. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Lencioni R: Loco-regional treatment of hepatocellular carcinoma. Hepatology. 52:762–773. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Llovet JM, Real MI, Montana X, Planas R, Coll S, Aponte J, Ayuso C, Sala M, Muchart J, Solà R, et al: Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: A randomised controlled trial. Lancet. 359:1734–1739. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Liu HC, Shan EB, Zhou L, Jin H, Cui PY, Tan Y and Lu YM: Combination of percutaneous radiofrequency ablation with transarterial chemoembolization for hepatocellular carcinoma: Observation of clinical effects. Chin J Cancer Res. 26:471–477. 2014.PubMed/NCBI

9 

European Association For The Study Of The Liver1; European Organisation For Research And Treatment Of Cancer, . EASL-EORTC clinical practice guidelines: Management of hepatocellular carcinoma. J Hepatol. 56:908–943. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Yu HC, Cheng JS, Lai KH, Lin CP, Lo GH, Lin CK, Hsu PI, Chan HH, Lo CC, Tsai WL and Chen WC: Factors for early tumor recurrence of single small hepatocellular carcinoma after percutaneous radiofrequency ablation therapy. World J Gastroenterol. 11:1439–1444. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Liao GS, Yu CY, Shih ML, Chan DC, Liu YC, Yu JC, Chen TW and Hsieh CB: Radiofrequency ablation after transarterial embolization as therapy for patients with unresectable hepatocellular carcinoma. Eur J Surg Oncol. 34:61–66. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Yan S, Xu D and Sun B: Combination of radiofrequency ablation with transarterial chemoembolization for hepatocellular carcinoma: A meta-analysis. Dig Dis Sci. 58:2107–2113. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Koh PS, Chan AC, Cheung TT, Chok KS, Dai WC, Poon RT and Lo CM: Efficacy of radiofrequency ablation compared with transarterial chemoembolization for the treatment of recurrent hepatocellular carcinoma: A comparative survival analysis. HPB (Oxford). Oct 16–2015.(Epub ahead of print). View Article : Google Scholar

14 

Tang C, Shen J, Feng W, Bao Y, Dong X, Dai Y, Zheng Y and Zhang J: Combination therapy of radiofrequency ablation and transarterial chemoembolization for unresectable hepatocellular carcinoma: A retrospective study. Medicine (Baltimore). 95:e37542016. View Article : Google Scholar : PubMed/NCBI

15 

Kokudo N, Hasegawa K, Akahane M, Igaki H, Izumi N, Ichida T, Uemoto S, Kaneko S, Kawasaki S, Ku Y, et al: Evidence-based clinical practice guidelines for hepatocellular carcinoma: The Japan Society of Hepatology 2013 update (3rd JSH-HCC Guidelines). Hepatol Res. 45:2015. View Article : Google Scholar

16 

Nishiofuku H, Tanaka T, Matsuoka M, Otsuji T, Anai H, Sueyoshi S, Inaba Y, Koyama F, Sho M, Nakajima Y and Kichikawa K: Transcatheter arterial chemoembolization using cisplatin powder mixed with degradable starch microspheres for colorectal liver metastases after FOLFOX failure: Results of a phase I/II study. J Vasc Interv Radiol. 24:56–65. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Bilehjani E and Fakhari S: Using central venous catheter for suprapubic catheterization in cardiac surgery. Res Rep Urol. 9:1–4. 2017.PubMed/NCBI

18 

Padhani AR and Ollivier L: The RECIST (Response Evaluation Criteria in Solid Tumors) criteria: Implications for diagnostic radiologists. Br J Radiol. 74:983–986. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Pagliaro L: MELD: The end of Child-Pugh classification? J Hepatol. 36:141–142. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Bruix J and Sherman M: Practice Guidelines Committee, American Association for the Study of Liver Diseases: Management of hepatocellular carcinoma. Hepatology. 42:1208–1236. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Jansen MC, van Hillegersberg R, Chamuleau RA, van Delden OM, Gouma DJ and van Gulik TM: Outcome of regional and local ablative therapies for hepatocellular carcinoma: A collective review. Eur J Surg Oncol. 31:331–347. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Yamashita Y, Torashima M, Oguni T, Yamamoto A, Harada M, Miyazaki T and Takahashi M: Liver parenchymal changes after transcatheter arterial embolization therapy for hepatoma: CT evaluation. Abdom Imaging. 18:352–356. 1993. View Article : Google Scholar : PubMed/NCBI

23 

Groupe d'Etude et de Traitement du Carcinome Hépatocellulaire: A comparison of lipiodol chemoembolization and conservative treatment for unresectable hepatocellular carcinoma. N Engl J Med. 332:1256–1261. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Wang X, Hu Y, Ren M, Lu X, Lu G and He S: Efficacy and safety of radiofrequency ablation combined with transcatheter arterial chemoembolization for hepatocellular carcinomas compared with radiofrequency ablation alone: A time-to-event meta-analysis. Korean J Radiol. 17:93–102. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Guo W, He X, Li Z and Li Y: Combination of transarterial chemoembolization (TACE) and radiofrequency ablation (RFA) vs. surgical resection (SR) on survival outcome of early hepatocellular carcinoma: A meta-analysis. Hepatogastroenterology. 62:710–714. 2015.PubMed/NCBI

26 

Chen QW, Ying HF, Gao S, Shen YH, Meng ZQ, Chen H, Chen Z and Teng WJ: Radiofrequency ablation plus chemoembolization versus radiofrequency ablation alone for hepatocellular carcinoma: A systematic review and meta-analysis. Clin Res Hepatol Gastroenterol. 40:309–314. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Jin YJ, Chung YH, Kim JA, Park W, Lee D, Shim JH, Lee D, Kim KM, Lim YS, Lee HC, et al: Predisposing factors of hepatocellular carcinoma recurrence following complete remission in response to transarterial chemoembolization. Dig Dis Sci. 58:1758–1765. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Kinugasa H, Nouso K, Takeuchi Y, Yasunaka T, Onishi H, Nakamura S, Shiraha H, Kuwaki K, Hagihara H, Ikeda F, et al: Risk factors for recurrence after transarterial chemoembolization for early-stage hepatocellular carcinoma. J Gastroenterol. 47:421–426. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Jeong SO, Kim EB, Jeong SW, Jang JY, Lee SH, Kim SG, Cha SW, Kim YS, Cho YD, Kim HS, et al: Predictive factors for complete response and recurrence after transarterial chemoembolization in hepatocellular carcinoma. Gut Liver. 11:409–416. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Park W, Chung YH, Kim JA, Jin YJ, Lee D, Shim JH, Lee D, Kim KM, Lim YS, Lee HC, et al: Recurrences of hepatocellular carcinoma following complete remission by transarterial chemoembolization or radiofrequency therapy: Focused on the recurrence patterns. Hepatol Res. 43:1304–1312. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Colecchia A, Schiumerini R, Cucchetti A, Cescon M, Taddia M, Marasco G and Festi D: Prognostic factors for hepatocellular carcinoma recurrence. World J Gastroenterol. 20:5935–5950. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Lee JK, Chung YH, Song BC, Shin JW, Choi WB, Yang SH, Yoon HK, Sung KB, Lee YS and Suh DJ: Recurrences of hepatocellular carcinoma following initial remission by transcatheter arterial chemoembolization. J Gastroenterol Hepatol. 17:52–58. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Matsuda M, Omata F, Fuwa S, Saida Y, Suzuki S, Uemura M, Ishii N, Iizuka Y, Fukuda K and Fujita Y: Prognosis of patients with hepatocellular carcinoma treated solely with transcatheter arterial chemoembolization: Risk factors for one-year recurrence and two-year mortality (preliminary data). Intern Med. 52:847–853. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Rou WS, Lee BS, Moon HS, Lee ES, Kim SH and Lee HY: Risk factors and therapeutic results of early local recurrence after transcatheter arterial chemoembolization. World J Gastroenterol. 20:6995–7004. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Matsumura H, Nirei K, Nakamura H, Higuchi T, Arakawa Y, Ogawa M, Tanaka N and Moriyama M: Histopathology of type C liver disease for determining hepatocellular carcinoma risk factors. World J Gastroenterol. 19:4887–4896. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Paradis V: Histopathology of hepatocellular carcinoma. Recent Results Cancer Res. 190:21–32. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Stefanini GF, Amorati P, Biselli M, Mucci F, Celi A, Arienti V, Roversi R, Rossi C, Re G and Gasbarrini G: Efficacy of transarterial targeted treatments on survival of patients with hepatocellular carcinoma. An Italian experience. Cancer. 75:2427–2434. 1995. View Article : Google Scholar : PubMed/NCBI

38 

Kwan SW, Fidelman N, Ma E, Kerlan RK Jr and Yao FY: Imaging predictors of the response to transarterial chemoembolization in patients with hepatocellular carcinoma: A radiological-pathological correlation. Liver Transpl. 18:727–736. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Sugimori K, Nozawa A, Morimoto M, Shirato K, Kokawa A, Saito T, Numata K and Tanaka K: Extension of radiofrequency ablation of the liver by transcatheter arterial embolization with iodized oil and gelatin sponge: Results in a pig model. J Vasc Interv Radiol. 16:849–856. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Seki T, Tamai T, Nakagawa T, Imamura M, Nishimura A, Yamashiki N, Ikeda K and Inoue K: Combination therapy with transcatheter arterial chemoembolization and percutaneous microwave coagulation therapy for hepatocellular carcinoma. Cancer. 89:1245–1251. 2000. View Article : Google Scholar : PubMed/NCBI

41 

Chang NK, Shin SS, Kim JW, Kim HJ, Jeong YY, Heo SH, Kim JK and Kang HK: Effect of ultrasound-guided radiofrequency ablation in incompletely treated hepatocellular carcinoma after transcatheter arterial chemoembolization. Korean J Radiol. 13 Suppl 1:S104–S111. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Xie H, Wang H, An W, Ma W, Qi R, Yang B, Liu C, Gao Y, Xu B and Wang W: The efficacy of radiofrequency ablation combined with transcatheter arterial chemoembolization for primary hepatocellular carcinoma in a cohort of 487 patients. PLoS One. 9:e890812014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 14 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Douhara A, Namisaki T, Moriya K, Kitade M, Kaji K, Kawaratani H, Takeda K, Okura Y, Takaya H, Noguchi R, Noguchi R, et al: Predisposing factors for hepatocellular carcinoma recurrence following initial remission after transcatheter arterial chemoembolization. Oncol Lett 14: 3028-3034, 2017
APA
Douhara, A., Namisaki, T., Moriya, K., Kitade, M., Kaji, K., Kawaratani, H. ... Yoshiji, H. (2017). Predisposing factors for hepatocellular carcinoma recurrence following initial remission after transcatheter arterial chemoembolization. Oncology Letters, 14, 3028-3034. https://doi.org/10.3892/ol.2017.6489
MLA
Douhara, A., Namisaki, T., Moriya, K., Kitade, M., Kaji, K., Kawaratani, H., Takeda, K., Okura, Y., Takaya, H., Noguchi, R., Nishimura, N., Seki, K., Sato, S., Sawada, Y., Yamao, J., Mitoro, A., Uejima, M., Mashitani, T., Shimozato, N., Saikawa, S., Nakanishi, K., Furukawa, M., Kubo, T., Yoshiji, H."Predisposing factors for hepatocellular carcinoma recurrence following initial remission after transcatheter arterial chemoembolization". Oncology Letters 14.3 (2017): 3028-3034.
Chicago
Douhara, A., Namisaki, T., Moriya, K., Kitade, M., Kaji, K., Kawaratani, H., Takeda, K., Okura, Y., Takaya, H., Noguchi, R., Nishimura, N., Seki, K., Sato, S., Sawada, Y., Yamao, J., Mitoro, A., Uejima, M., Mashitani, T., Shimozato, N., Saikawa, S., Nakanishi, K., Furukawa, M., Kubo, T., Yoshiji, H."Predisposing factors for hepatocellular carcinoma recurrence following initial remission after transcatheter arterial chemoembolization". Oncology Letters 14, no. 3 (2017): 3028-3034. https://doi.org/10.3892/ol.2017.6489