Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Combined therapy of transarterial chemoembolization and stereotactic body radiation therapy versus transarterial chemoembolization for ≤5cm hepatocellular carcinoma: Propensity score matching analysis

  • Baek Gyu Jun ,

    Contributed equally to this work with: Baek Gyu Jun, Sang Gyune Kim

    Roles Conceptualization, Data curation, Writing – original draft

    Affiliation Department of Internal Medicine, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung, South Korea

  • Sang Gyune Kim ,

    Contributed equally to this work with: Baek Gyu Jun, Sang Gyune Kim

    Roles Methodology, Writing – review & editing

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Bucheon Hospital, Bucheon, South Korea

  • Young Don Kim,

    Roles Data curation

    Affiliation Department of Internal Medicine, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung, South Korea

  • Gab Jin Cheon,

    Roles Validation

    Affiliation Department of Internal Medicine, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung, South Korea

  • Koon Hee Han,

    Roles Data curation

    Affiliation Department of Internal Medicine, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung, South Korea

  • Jeong-Ju Yoo,

    Roles Data curation

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Bucheon Hospital, Bucheon, South Korea

  • Young Seok Kim,

    Roles Validation

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Bucheon Hospital, Bucheon, South Korea

  • Soung Won Jeong,

    Roles Supervision

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Seoul Hospital, Seoul, South Korea

  • Jae Young Jang,

    Roles Supervision

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Seoul Hospital, Seoul, South Korea

  • Sae Hwan Lee,

    Roles Data curation

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan, South Korea

  • Suyeon Park,

    Roles Methodology

    Affiliation Department of Biostatistics, College of Medicine, Soonchunhyang University, Seoul, Korea

  • Hong Soo Kim

    Roles Writing – review & editing

    khskhs@schmc.ac.kr

    Affiliation Department of Internal Medicine, Soonchunhyang University College of Medicine Cheonan Hospital, Cheonan, South Korea

Abstract

Patients with liver cirrhosis and hepatocellular carcinoma (HCC) are often ineligible for resection or local ablation therapy due to poor liver function and/or difficult location. The aim of this study is to evaluate therapeutic outcomes of stereotactic body radiotherapy (SBRT) combined with transarterial chemoembolization (TACE) compared with TACE alone for HCC measuring less than 5 cm. From March 2011 to December 2016, 85 patients underwent SBRT with TACE (SBRT-TACE group) and 114 underwent TACE (TACE group) at 4 tertiary hospitals. Local control rate (LCR), progression-free survival (PFS) and overall survival (OS) were compared after propensity-score matching (1:1 ratio). The SBRT-TACE group showed significantly higher 1- and 3-year LCR than the TACE group (91.1% and 89.9%, respectively vs 69.9% and 44.8%, respectively; P < 0.001). The SBRT-TACE group showed better 1- and 3-year PFS than the TACE group (56.5% and 32.3%, respectively vs 42.2% and 21.6%, respectively; P = 0.022). However, 1-, 3- and 5-year OS was not different between the SBRT-TACE and TACE groups (98.8%, 89.1% and 80.7%, respectively vs 99.7%, 83.3% and 71.0%, respectively; P = 0.206). In multivariate analysis, the overall SBRT added to TACE did not contribute to extend PFS. However, in patients with less than 2 tumors, the combined therapy was effective (HR 0.590, 95% CI 0.392–0.889, P = 0.012). SBRT-TACE is superior to TACE in terms of LCR. Particularly, SBRT-TACE may be an effective alternative in patients with HCC number (≤2), which is not indicated for resection or local ablation.

Introduction

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death worldwide [1]. Resection is the standard treatment for early-stage HCC [2]. However, many patients are not indicated for resection or ablative therapy because of advanced cirrhosis or tumor location [3]. Surgery is not indicated for elderly patients in poor general condition. Transarterial chemoembolization (TACE) is usually performed as an alternative treatment [4, 5]. The effect of TACE is well established especially in patients with Barcelona clinic liver cancer (BCLC) stage B. Unfortunately, the response rate of conventional TACE is relatively low (40%), and therefore, regarded as a palliative treatment [6, 7].

Traditionally, radiotherapy (RT) played a limited role due to radiation-induced liver disease (RILD) [8]. Recently, stereotactic body radiotherapy (SBRT) has emerged as a new modality of HCC treatment. Technological advances allow RT using high doses of radiation to conform to the target volume safely [9, 10]. Compared with conventional RT as a palliative approach, which is associated with low local control (LC), stereotactic body radiotherapy (SBRT) results in a high rate of LC, by delivering a high dose of radiation in a few fractions to small HCC [11]. Further, adjuvant SBRT following TACE is an effective treatment modality in relatively medium-sized HCC [12]. With advances in radiation technology, RILD after SBRT treatment was tolerable even in patients with Child-Pugh (CP) score ≤7 [13].

However, the efficacy of SBRT combined with TACE compared with TACE alone is unknown. In this study, we investigated the effect of SBRT and TACE combination versus TACE alone on tumor response and patient survival.

Methods

Patients

Data of HCC patients who underwent TACE as an initial treatment between March 2011 and February 2016 were reviewed at four tertiary referral hospitals (Soonchunhyang University Seoul, Bucheon, Cheonan Hospital, and Gangneung Asan Medical Center). Patients following the inclusion criteria were selected: 1) tumor size ≤5 cm of long diameter, and ≤3 lesions present; 2) ineligible for resection or local ablative therapies; and 3) CP class A or B. The exclusion criteria were as follows: 1) previous treatment of resection or radiofrequency ablation or TACE; 2) extrahepatic metastasis; and 3) presence of vascular invasion or portal vein tumor thrombosis. The diagnosis of HCC was made by using dynamic imaging technique [14].

One hundred fourteen patients were treated with TACE alone (TACE group) while eighty five patients were treated with TACE in combination with SBRT (TACE-SBRT group). The selection criteria for TACE and SBRT were mainly determined by considering tumor vascularity, hepatic angiography, accessibility, risk of bleeding or liver toxicity. We conducted propensity score matching to minimize the differences in underlying confounding factors between the two groups (1:1 ratio). This study was approved by the Institutional Review Board of Soonchunhyang University Seoul, Bucheon, Cheonan Hospital, and Gangneung Asan Medical Center, and written informed consent was waived because of the retrospective study.

TACE

TACE was performed via the common femoral artery using an angiographic catheter followed by selection of feeder vessels of hepatic segments. Patients were treated with a mixture of intra-arterial adriamycin (50 mg/m2) and lipiodol (5 to 10 mL) with gelfoam embolization at Soonchunhyang University Seoul, Cheoan, Bucheon Hospital and Gangeung Asan Hospital [15, 16].

SBRT

SBRT was carried out at Soonchunhyang University Seoul Hospital using the CyberKnife Radiosurgery System (Accuray Incorporated, Sunnyvale, CA), Soonchunhyang University Cheonan using the Novalis TX (Varian Medical Systems and BrainLab), Soonchunhyang University Bucheon using the TomoTherapy device (Madison, WI, USA) and Gangneung Asan Medical Center using the TrueBeam medical linear accelerator (Varian Medical Systems, Palo Alto, CA, USA). SBRT was performed viable tumors that showed incomplete response after first TACE based on follow-up CT. Patients were immobilized in supine position with arms above their head. Gross tumor volume (GTV) was measured based on CT images at the end-expiratory phase fused with multi-phase MR images. Extension based on movement within the gating phase (30–70%) from the GTV was set as the internal target volume (ITV). The planning target volume (PTV) was defined as the volume with a 5 mm margin added to the ITV. A total dose of 40–60 Gy (median, 55 Gy) was administered in the PTV of three to five fractions over consecutive days or twice a week [13].

Liver toxicity

Liver toxicity was defined as worsening of CP score by 2 or more within 3 months or elevated liver transaminases more than five times the upper normal limit after treatment [17].

Assessment

The primary endpoint included comparison of the overall survival (OS) in the SBRT-TACE and TACE groups. The secondary endpoint was the comparison of LC and progression-free survival (PFS).

All patients were followed up every 1 to 3 months. Physical examinations, complete blood cell counts, biochemical profiles, tumor markers, and three-phasic CT or magnetic resonance imaging (MRI) scans were performed at every follow-up visit. Complications were assessed according to version 4 of the Common Terminology Criteria for Adverse Events. Liver toxicity was defined as elevated liver transaminases more than five times the upper normal limit or worsening of CP score by 2 or more within 3 months after SBRT [17].

Statistical analysis

The OS, PFS and LC in each treatment group were estimated using the Kaplan-Meier method and log-rank test. OS was calculated from the date of diagnosis until the date of final follow-up or death. PFS was estimated from the date of initial TACE until the date of extra- and/or intrahepatic disease progression, recurrence, or death. LC was defined as the absence of progressive disease (PD) within the PTV as per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 in multiphasic CT or MRI. Lesions that developed or progressed outside the PTV in the liver or lymph nodes were scored as regional PD and those developed in other organs as distant PD. Survival and control times were calculated from the start of SBRT. Time to progression and survival were evaluated with the Kaplan-Meier method [18]. Cox proportional-hazards model was used to evaluate the factors influencing PFS and OS rates.

To reduce the effect of potential confounding in a retrospective study, we also performed rigorous adjustment for differences in baseline characteristics of patients using propensity score methods (R version 3.1.2, ‘MatchIt’ package). The SBRT-TACE and TACE groups were matched 1:1 to maximize the propensity score match. Age, gender, tumor size, number of tumors, Child-Pugh score, and BCLC stage were selected on the basis of this score, and calculated from baseline characteristics. A P-value of < 0.05 was considered significant. All statistical analyses were performed using SPSS statistical package (version 18.0; SPSS Inc., Chicago, IL, USA).

Results

Patient characteristics before and after propensity score matching

After propensity score matching at a 1:1 ratio, the SBRT-TACE and TACE groups comprised 85 patients, respectively. No significant differences in sex (P = 0.858), age (P = 0.894), tumor number (P = 0.816), tumor size (P = 0.753), etiology (P = 0.778) and CP score (P = 0.663) were observed (Table 1).

thumbnail
Table 1. Baseline characteristics before and after propensity score matching.

https://doi.org/10.1371/journal.pone.0206381.t001

Local control, progression-free survival and overall survival after propensity score matching

The SBRT-TACE group showed significantly higher 1-, 3- and 5-year LC rates than the TACE group (91.1%, 89.9% and 89.9%, respectively vs. 69.9%, 44.8% and 44.8%, respectively; P < 0.001) (Fig 1). The SBRT-TACE group showed better 1- and 3- year PFS than the TACE groups (56.5% and 32.3%, respectively vs. 42.2% and 21.6%, respectively; P = 0.022) (Fig 2). However, 1-, 3- and 5-year OS was not different between the SBRT+ TACE and TACE groups (98.8%, 89.1% and 80.7%, respectively vs. 99.7%, 83.3% and 71.0%, respectively; P = 0.206) (Fig 3).

thumbnail
Fig 1. Comparison of the local control rates between SBRT-TACE and TACE groups.

(p<0.001).

https://doi.org/10.1371/journal.pone.0206381.g001

thumbnail
Fig 2. Comparison of progression-free survival rates between SBRT-TACE and TACE groups.

(p = 0.022).

https://doi.org/10.1371/journal.pone.0206381.g002

thumbnail
Fig 3. Comparison of overall survival rates between SBRT-TACE and TACE groups.

(P = 0.206).

https://doi.org/10.1371/journal.pone.0206381.g003

Prognostic factors for progression-free survival and overall survival after propensity score matching

In multivariate analysis, BCLC stage (stage B) (hazard ratio [HR] = 3.701, 95% confidence interval [CI] 1.635–8.379, P = 0.002), number of tumors (n≥3) (HR = 2.710, 95% CI 1.494–4.915, P < 0.001) and CP class B (vs. A) (HR = 1.945, 95% CI 1.183–3.197, P = 0.009) were associated with poor PFS. After adjusting for other variables, SBRT-TACE showed a marginal trend toward significance (HR = 0.695, 95% CI 0.480–1.005, P = 0.053) (Table 2).

thumbnail
Table 2. Prognostic factors for progression-free survival after propensity score matching.

https://doi.org/10.1371/journal.pone.0206381.t002

CP class B (vs. A) (HR = 2.570, 95% CI 1.241–5.324, P = 0.011) and BLCL stage (stage B) (HR = 5.835, 95% CI 1.719–19.801, P = 0.05) were significantly poor prognostic factors for OS. Tumor size (HR = 1.179, 95% CI 0.746–1.863, P = 0.482) and tumor number (n≤2: HR = 0.582, 95% CI 0.219–1.548, P = 0.278) (n≥3: HR = 1.697, 95% CI 0.652–4.417, P = 0.279) were not associated with OS (Table 3).

thumbnail
Table 3. Prognostic factors for overall survival after propensity score matching.

https://doi.org/10.1371/journal.pone.0206381.t003

Subgroup analysis by tumor number

Based on previous studies, SBRT was more effective in patients with a small number of HCCs. In our patients with less than two HCCs, the SBRT-TACE group showed better 1- and 3- year PFS than TACE groups (61.0% and 42.2%, respectively vs. 47.6% and 21.6%, respectively; P = 0.006) (Fig 4). SBRT-TACE group (HR = 0.590, 95% CI 0.392–0.889, P = 0.012) showed a significantly increased PFS after adjustment for BCLC stage tumor size and CP class if a patient had 1 or 2 HCC nodules (Table 4).

thumbnail
Fig 4. Comparison of progression-free survival with SBRT-TACE and TACE in subgroup analysis by tumor number.

(n≤2) (p = 0.006).

https://doi.org/10.1371/journal.pone.0206381.g004

thumbnail
Table 4. Prognostic factors for progression-free survival in patients with 1 to 2 nodules after propensity score matching.

https://doi.org/10.1371/journal.pone.0206381.t004

Liver toxicity

SBRT-TACE and TACE groups showed no difference in liver toxicity after treatment. Worsening of CP score by 2 or more within 3 months after treatment occurred in 8 out of 85 (9.4%) in the SBRT-TACE group and 3 out of 85 (5.5%) in the TACE group, respectively (P = 0.119). Elevated liver transaminases more than five times the upper normal limit after treatment occurred in 8 out of 85 (9.4%) in the SBRT-TACE group and 4 out of 85 (4.8%) in the TACE group, respectively (P = 0.239).

Discussion

Only 30–40% of HCC patients undergo curative treatment because many patients with early-stage HCC indicated for resection or local ablation already have advanced liver cirrhosis [19]. SBRT is an emerging technique for patients who are not indicated for radical therapy [20]. In this study, we compared the therapeutic outcome of SBRT-TACE and TACE with propensity score matching. The combination therapy of SBRT and TACE may be more effective than TACE in terms of LC rate. Furthermore, in subgroup analysis, SBRT-TACE in patients with HCCs less than 2 resulted in better PFS without increased liver toxicity. SBRT-TACE represents a favorable alternative for treatment of patients with HCCs less than 2.

As reported in most published clinical studies, SBRT was associated with a favorable LC rate [11, 21, 22]. However, complete tumor response after TACE alone was a challenge [6, 7]. In our study, we reported high LC rate (89.9% at 3 years) in the SBRT-TACE group compared with that of TACE group (44.8% at 3 years). Especially, SBRT resulted in high LC rates in patients with small HCC similar to previous studies. Yoon et al. have reported that LC rate at 3 years was 100% in patients with tumors ≤ 2 cm, and 93.3% in patients with tumors between 2.1–3 cm [11]. Takeda et al also have shown high local control rate (96.3% at 3 years) in HCC (≤ 4cm) patients.[21] In a comparative study of SBRT versus radiofrequency ablation (RFA), similar LC rates were found in patients with small HCC [23]. However, the LC rate of SBRT was not satisfactory with increased tumor size [24, 25].

TACE combined with SBRT has been reported to be effective and safe for the treatment of small- or medium-sized HCC [12, 26, 27]. Jacob et al reported that in patients with HCC tumors measuring ≥3 cm, treatment with TACE-SBRT significantly decreased local recurrence in comparison with TACE alone[26]. In a study from Japan, complete response to therapy was noted in 29 (96.3%) patients belonging to the SBRT-TACE group and in only one (3.3%) patient included in the TACE group (P < 0.001) [27]. In a prospective study, SBRT-TACE showed a promising LC rate in HCC (<10 cm) [12]. We also suggest that SBRT-TACE is an effective treatment for both small- and medium-sized HCC with a high LC rate.

In the current study, we achieved better PFS in the SBRT-TACE group than in the TACE group. However, in multivariate analysis, SBRT-TACE was not a predictive factor for PFS. Tumor number (n≥3), CP class B and BLCL stage B were associated with worse PFS (Table 2). According to a previous study, multiple HCC nodules represented an important prognostic factor in PFS [25]. We performed subgroup analysis with HCC number less than 2 and found that SBRT-TACE was a significant prognostic factor of longer PFS (Table 4). In a study, which was limited to patients with HCC carrying 1 to 2 concurrent liver tumors, SBRT resulted in a better PFS than TACE (P < 0.001) and TACE was associated with worse PFS in multivariate analysis (HR 3.35, P < 0.01) [28].

The effect of SBRT on survival is disputed. A randomized controlled study investigating the efficacy of SBRT and SBRT with TACE has never been conducted. Previous studies reported that OS of SBRT was not inferior to OS following curative treatment. In retrospective studies, OS of SBRT was similar to that of RFA [23, 29]. Su et al suggested that SBRT and liver resection provide similar 5-year OS for small HCC (74.3 vs 69.2%, P = 0.405) [30]. However, in this study we did not show that OS of SBRT-TACE was better than that of TACE alone (P = 0.206) (Fig 3). Multivariate analysis showed that SBRT-TACE did not increase OS (HR 0.722, P = 0.324) (Table 3). Similar to our study, Sapir et al reported no difference in OS between patients treated with TACE or SBRT after propensity score matching [28]. This study did not show difference in OS between the two groups. Possible reasons for such results are described below. First, various treatment modalities were performed for recurrence after TACE only or TACE+SBRT. Second, baseline liver function was similar after propensity score matching. A retrospective study has shown that long-term survival rates after TACE are comparable to those after resection and RFA for small single-nodule HCC. That study explained that degree of baseline liver dysfunction was more important than specific treatment modality itself [31]. Therefore, a prospective controlled trial comparing SBRT-TACE and TACE is warranted to elucidate the survival effect of SBRT-TACE.

We analyzed the differences in liver toxicity after SBRT-TACE or TACE. The incidence of worsening CP score or increased transaminase levels was slightly high in SBRT-TACE group. However, it did not show statistical difference. Many clinical studies have reported that patients did not experience severe radiation-induced liver damage after combined SBRT and TACE [27, 32]. Therefore, SBRT-TACE is a safe option for patients with small HCCs.

This study has a few limitations. First, this study is retrospective. However, with propensity score matching adjusting for potential confounders, this study comparing the benefits of SBRT-TACE with those of TACE was well-balanced. Second, this study is a multicenter study, with variation in devices across multiple institutions. All procedures were performed by the same operator at individual hospitals. However, there was no difference in the treatment methods and response evaluation of HCC between in the four hospitals.

Conclusion

This study showed that SBRT-TACE compared with TACE is a feasible option for patients with HCC (≤5cm) without increased liver toxicity. SBRT-TACE increased LC rate. We suggest that the advantages of SBRT-TACE should be demonstrated in patients with small HCC. SBRT-TACE is superior to TACE in terms of LCR. Particularly, SBRT-TACE has better PFS than TACE in patients with HCC number (≤2). SBRT-TACE represents an alternative treatment modality.

References

  1. 1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010; 127: 2893–2917. https://doi.org/10.1002/ijc.25516 pmid:21351269
  2. 2. European Association for the Study of the Liver, European Organisation for Research, Treatment of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012; 56: 908–943. https://doi.org/10.1016/j.jhep.2011.12.001 pmid:22424438
  3. 3. Ikai I, Kudo M, Arii S, Omata M, Kojiro M, Sakamoto M, et al. Report of the 18th follow‐up survey of primary liver cancer in Japan. Hepatol Res. 2010; 40: 1043–1059. https://doi.org/10.1111/j.1872-034X.2010.00731.x
  4. 4. Sheu JC, Huang GT, Chen DS, Sung JL, Yang PM, Wei TC, et al. Small hepatocellular carcinoma: intratumor ethanol treatment using new needle and guidance systems. Radiology. 1987; 163: 43–48. https://doi.org/10.1148/radiology.163.1.3029806 pmid:3029806
  5. 5. Ikeda K, Kumada H, Saitoh S, Arase Y, Chayama K. Effect of repeated transcatheter arterial embolization on the survival time in patients with hepatocellular carcinoma. An analysis by the Cox proportional hazard model. Cancer. 1991; 68: 2150–2154. pmid:1655202
  6. 6. Maluccio MA, Covey AM, Porat LB, Schubert J, Brody LA, Sofocleous CT, et al. Transcatheter arterial embolization with only particles for the treatment of unresectable hepatocellular carcinoma. J Vasc Interv Radiol. 2008; 19: 862–869. https://doi.org/10.1016/j.jvir.2008.02.013 pmid:18503900
  7. 7. Terzi E, Piscaglia F, Forlani L, Mosconi C, Renzulli M, Bolondi L, et al. TACE performed in patients with a single nodule of hepatocellular carcinoma. BMC Cancer. 2014; 14: 601. https://doi.org/10.1186/1471-2407-14-601 pmid:25139639
  8. 8. Lawrence TS, Robertson JM, Anscher MS, Jirtle RL, Ensminger WD, Fajardo LF. Hepatic toxicity resulting from cancer treatment. Int J Radiat Oncol Biol Phys. 1995; 31: 1237–1248. https://doi.org/10.1016/0360-3016(94)00418-K pmid:7713785
  9. 9. Dawson LA, Normolle D, Balter JM, McGinn CJ, Lawrence TS, Ten Haken RK. Analysis of radiation-induced liver disease using the Lyman NTCP model. Int J Radiat Oncol Biol Phys. 2002; 53: 810–821. pmid:12095546
  10. 10. Liang SX, Zhu XD, Xu ZY, Zhu J, Zhao JD, Lu HJ, et al. Radiation-induced liver disease in three-dimensional conformal radiation therapy for primary liver carcinoma: the risk factors and hepatic radiation tolerance. Int J Radiat Oncol Biol Phys. 2006; 65: 426–434. https://doi.org/10.1016/j.ijrobp.2005.12.031 pmid:16690430
  11. 11. Yoon SM, Lim YS, Park MJ, Kim SY, Cho B, Shim JH, et al. Stereotactic body radiation therapy as an alternative treatment for small hepatocellular carcinoma. PLoS One. 2013; 8: e79854. https://doi.org/10.1371/journal.pone.0079854 pmid:24255719
  12. 12. Kang JK, Kim MS, Cho CK, Yang KM, Yoo HJ, Kim JH, et al. Stereotactic body radiation therapy for inoperable hepatocellular carcinoma as a local salvage treatment after incomplete transarterial chemoembolization. Cancer. 2012; 118: 5424–5431. https://doi.org/10.1002/cncr.27533 pmid:22570179
  13. 13. Jun BG, Kim YD, Cheon GJ, Kim ES, Jwa E, Kim SG, et al. Clinical significance of radiation-induced liver disease after stereotactic body radiation therapy for hepatocellular carcinoma. Korean J Intern Med. 2017. https://doi.org/10.3904/kjim.2016.412 pmid:28844122
  14. 14. Bruix J, Sherman M, American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011; 53: 1020–1022. https://doi.org/10.1002/hep.24199 pmid:21374666
  15. 15. Jeong SO, Kim EB, Jeong SW, Jang JY, Lee SH, Kim SG, et al. Predictive factors for complete response and recurrence after transarterial chemoembolization in hepatocellular carcinoma. Gut Liver. 2017; 11: 409–416. https://doi.org/10.5009/gnl16001 pmid:28208001
  16. 16. Kim MJ, Kim YS, Cho YH, Jang HY, Song JY, Lee SH, et al. Use of (18)F-FDG PET to predict tumor progression and survival in patients with intermediate hepatocellular carcinoma treated by transarterial chemoembolization. Korean J Intern Med. 2015; 30: 308–315. https://doi.org/10.3904/kjim.2015.30.3.308 pmid:25995661
  17. 17. Pan CC, Kavanagh BD, Dawson LA, Li XA, Das SK, Miften M, et al. Radiation-associated liver injury. Int J Radiat Oncol Biol Phys. 2010; 76: S94–100. https://doi.org/10.1016/j.ijrobp.2009.06.092 pmid:20171524
  18. 18. Gkika E, Schultheiss M, Bettinger D, Maruschke L, Neeff HP, Schulenburg M, et al. Excellent local control and tolerance profile after stereotactic body radiotherapy of advanced hepatocellular carcinoma. Radiat Oncol. 2017; 12: 116. https://doi.org/10.1186/s13014-017-0851-7 pmid:28701219
  19. 19. 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. https://doi.org/10.1002/hep.510300629 pmid:10573522
  20. 20. Sanuki N, Takeda A, Kunieda E. Role of stereotactic body radiation therapy for hepatocellular carcinoma. World J Gastroenterol. 2014; 20: 3100–3111. https://doi.org/10.3748/wjg.v20.i12.3100 pmid:24696597
  21. 21. Takeda A, Sanuki N, Tsurugai Y, Iwabuchi S, Matsunaga K, Ebinuma H, et al. Phase 2 study of stereotactic body radiotherapy and optional transarterial chemoembolization for solitary hepatocellular carcinoma not amenable to resection and radiofrequency ablation. Cancer. 2016; 122: 2041–2049. https://doi.org/10.1002/cncr.30008 pmid:27062278
  22. 22. Sanuki N, Takeda A, Oku Y, Mizuno T, Aoki Y, Eriguchi T, et al. Stereotactic body radiotherapy for small hepatocellular carcinoma: a retrospective outcome analysis in 185 patients. Acta Oncol. 2014; 53: 399–404. https://doi.org/10.3109/0284186X.2013.820342 pmid:23962244
  23. 23. Wahl DR, Stenmark MH, Tao Y, Pollom EL, Caoili EM, Lawrence TS, et al. Outcomes after stereotactic body radiotherapy or radiofrequency ablation for hepatocellular carcinoma. J Clin Oncol. 2016; 34: 452–459. https://doi.org/10.1200/JCO.2015.61.4925 pmid:26628466
  24. 24. Que JY, Lin LC, Lin KL, Lin CH, Lin YW, Yang CC. The efficacy of stereotactic body radiation therapy on huge hepatocellular carcinoma unsuitable for other local modalities. Radiat Oncol. 2014; 9: 120. https://doi.org/10.1186/1748-717X-9-120 pmid:24885086
  25. 25. Su TS, Liang P, Lu HZ, Liang J, Gao YC, Zhou Y, et al. Stereotactic body radiation therapy for small primary or recurrent hepatocellular carcinoma in 132 Chinese patients. J Surg Oncol. 2016; 113: 181–187. https://doi.org/10.1002/jso.24128 pmid:26799260
  26. 26. Jacob R, Turley F, Redden DT, Saddekni S, Aal AK, Keene K, et al. Adjuvant stereotactic body radiotherapy following transarterial chemoembolization in patients with non-resectable hepatocellular carcinoma tumours of >/ = 3 cm. HPB (Oxford). 2015; 17: 140–149. https://doi.org/10.1111/hpb.12331 pmid:25186290
  27. 27. Honda Y, Kimura T, Aikata H, Kobayashi T, Fukuhara T, Masaki K, et al. Stereotactic body radiation therapy combined with transcatheter arterial chemoembolization for small hepatocellular carcinoma. J Gastroenterol Hepatol. 2013; 28: 530–536. https://doi.org/10.1111/jgh.12087 pmid:23216217
  28. 28. Sapir E, Tao Y, Schipper MJ, Bazzi L, Novelli PM, Devlin P, et al. Stereotactic body radiation therapy as an alternative to transarterial chemoembolization for hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2018; 100: 122–130. https://doi.org/10.1016/j.ijrobp.2017.09.001 pmid:29066120
  29. 29. Seo YS, Kim MS, Yoo HJ, Jang WI, Paik EK, Han CJ, et al. Radiofrequency ablation versus stereotactic body radiotherapy for small hepatocellular carcinoma: a Markov model-based analysis. Cancer Med. 2016; 5: 3094–3101. https://doi.org/10.1002/cam4.893 pmid:27709795
  30. 30. Su TS, Liang P, Liang J, Lu HZ, Jiang HY, Cheng T, et al. Long-term survival analysis of stereotactic ablative radiotherapy versus liver resection for small hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2017; 98: 639–646. https://doi.org/10.1016/j.ijrobp.2017.02.095 pmid:28581406
  31. 31. Yang HJ, Lee JH, Lee DH, Yu SJ, Kim YJ, Yoon JH, et al. Small single-nodule hepatocellular carcinoma: comparison of transarterial chemoembolization, radiofrequency ablation, and hepatic resection by using inverse probability weighting. Radiology. 2014; 271: 909–918. https://doi.org/10.1148/radiol.13131760 pmid:24520944
  32. 32. Su TS, Lu HZ, Cheng T, Zhou Y, Huang Y, Gao YC, et al. Long-term survival analysis in combined transarterial embolization and stereotactic body radiation therapy versus stereotactic body radiation monotherapy for unresectable hepatocellular carcinoma >5 cm. BMC Cancer. 2016; 16: 834. https://doi.org/10.1186/s12885-016-2894-9 pmid:27809890